EP2010571B1 - Humanized c-kit antibody - Google Patents

Humanized c-kit antibody Download PDF

Info

Publication number
EP2010571B1
EP2010571B1 EP07756072.0A EP07756072A EP2010571B1 EP 2010571 B1 EP2010571 B1 EP 2010571B1 EP 07756072 A EP07756072 A EP 07756072A EP 2010571 B1 EP2010571 B1 EP 2010571B1
Authority
EP
European Patent Office
Prior art keywords
antibody
amino acid
kit
cells
acid sequence
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
EP07756072.0A
Other languages
German (de)
English (en)
French (fr)
Other versions
EP2010571A2 (en
Inventor
Gordon Ng
Wenyan Shen
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Amgen Inc
Original Assignee
Amgen Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Amgen Inc filed Critical Amgen Inc
Priority to EP18181319.7A priority Critical patent/EP3453724A1/en
Publication of EP2010571A2 publication Critical patent/EP2010571A2/en
Application granted granted Critical
Publication of EP2010571B1 publication Critical patent/EP2010571B1/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/08Drugs for disorders of the alimentary tract or the digestive system for nausea, cinetosis or vertigo; Antiemetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/12Antidiarrhoeals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/04Drugs for skeletal disorders for non-specific disorders of the connective tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/06Antimigraine agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/12Ophthalmic agents for cataracts
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/04Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/40Immunoglobulins specific features characterized by post-translational modification
    • C07K2317/41Glycosylation, sialylation, or fucosylation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/71Decreased effector function due to an Fc-modification
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/77Internalization into the cell
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • This invention relates to compositions for treating c-Kit associated inflammatory, fibrotic, autoimmune and cancerous diseases and to compositions containing humanized c-Kit antibodies.
  • Mast cells have been implicated in the mediation of inflammatory conditions such as asthma, rheumatoid arthritis and inflammatory bowel disease and the role in allergic inflammation is widely recognized.
  • Mast cells are increased in number in lung explants from severe asthmatics and are the major source of clinically relevant inflammatory mediators such as leukotriences, histamine and Th2 cytokines.
  • Mast cells are the major source of pre-formed TNF in disease tissues.
  • SCF Stem Cell Factor
  • c-Kit is a glycoprotein that signals through the cell and membrane associated tyrosine kinase receptor hereafter defined as c-Kit, and this signaling pathway plays a key role in hematopoiesis acting both as a positive and negative regulator, often in synergy with other cytokines.
  • a soluble shed c-Kit receptor may play a role in regulating SCF.
  • C-Kit is expressed on pluripotent hematopoietic stem cells which are the precurors to mature cells belonging to lymphoid and erythroid lineages. Unlike other hematopoietic cells, mast cell precursors and mature mast cells retain high-levels of c-Kit expression.
  • SCF signaling via c-Kit is vital for mast cell development, function, trafficking and survival. It also plays a role in gametogenesis, and melanogenesis. Mice with inactivating c-Kit mutations in the W locus have virtually no mast cells. Activating c-Kit mutations in man are associated with mastocytosis.
  • c-Kit positive pluripotent hematopoietic stem cells are precursors to multiple cell types including mesenchymal cells, fibroblasts and mast cells. Fibrotic disease is characterized in part by excessive fibroblast activity and proliferation resulting in the extracellular matrix deposition. C-Kit positive bone marrow pluripotent hematopoietic stem cells have been reported to be a source of the fibroblasts and mast cells in fibrotic tissues.
  • Mast cells can provide a sustained source of inflammatory, angiogenic, mitogenic and fibrogenic mediators.
  • Mast cells are functionally and anatomically coupled to fibroblasts and have a direct role in activating fibroblasts.
  • Mast cells increase the kinetics and magnitude of fibroblast mediated collagen contraction, extracellular matrix deposition and can transform fibroblasts into myofibroblasts.
  • Fibroblasts in turn secrete SCF to further activate and expand mast cells, and both cell types are components of the fibrogenic network.
  • mast cell number and mast cell mediators are significantly elevated in most human fibrotic diseases including idiopathic pulmonary fibrosis (IPF) and Scleroderma. Differential mast cell phenotypes are detected in some scleroderma patients and in the Tsk mouse model of scleroderma. An aggressive systemic form of mastocytosis may be characterized by myelofibrosis indicating that mast cells can be effector cells in fibrosis.
  • IPF idiopathic pulmonary fibrosis
  • Scleroderma Differential mast cell phenotypes are detected in some scleroderma patients and in the Tsk mouse model of scleroderma.
  • An aggressive systemic form of mastocytosis may be characterized by myelofibrosis indicating that mast cells can be effector cells in fibrosis.
  • GleevecTM and others in the class like SutentTM are multi-targeted tyrosine kinase receptor inhibitors that can target c-Kit signaling activity, but inhibit a number of other kinases. These kinase inhibitors are indicated for the oncology setting. Myelosuppression, anemia and a number of side-effects including cardiotoxicity and peripheral edema have been reported for Gleevec. Therefore these molecules may not possess the best benefit to risk profile for chronic treatment of diseases associated with c-Kit signaling. Thus, there is a need for new therapies and reagents, particularly those that are more potent and selective, and possess better safety profiles for the treatment of c-Kit associated inflammatory and fibrotic diseases.
  • Such a compound could also show significantly better efficacy and safety profiles in oncologic diseases such as myeloid derived leukemia, diseases associated with c-Kit mutations such as GIST and mastocytosis, diseases associated with over-expression of c-kit and/or excessive SCF autocrine activity as in melanoma and various SCLCs.
  • oncologic diseases such as myeloid derived leukemia, diseases associated with c-Kit mutations such as GIST and mastocytosis, diseases associated with over-expression of c-kit and/or excessive SCF autocrine activity as in melanoma and various SCLCs.
  • therapies and reagents targeting human c-Kit and capable of affecting a therapeutic benefit without significant adverse effects are currently lacking.
  • the invention provides agents that are antagonists and neutral antagonists of SCF at the cell-associated and membrane c-Kit receptor, such as monoclonal antibodies.
  • humanized (non-murine) monoclonal antibodies that bind c-Kit are provided.
  • the humanized antibodies of the invention comprise an amino acid sequence selected from those set forth in SEQ ID NOs 2 and 4.
  • nucleic acids encoding any of the preceding antibodies or specific binding agents.
  • a vector comprising any of the aforementioned nucleic acid sequences is provided.
  • a host cell is provided comprising any of the aforementioned nucleic acids or vectors.
  • the c-Kit binding agents and neutral antagonists are aglycosylated IgG1 antibodies that specifically bind c-Kit and that comprise an amino acid sequence of SEQ ID NO: 4 and an amino acid sequence that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more identical to the amino acid sequence set forth in SEQ ID NO: 2; or an amino acid sequence of SEQ ID NO: 2 and an amino acid sequence that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more identical to the amino acid sequence set forth in SEQ ID NO: 4.
  • sequence variation relative to SEQ ID NO: 2or 4 may represent, for example, a conservative substitution of the corresponding framework region of an IgG using an alternative human amino acid at that position.
  • the aforementioned antibodies exhibit an avidity characterized by a k d of lower than 1 x 10 -8 , or 1 x 10 -9 , as determined by surface plasmon resonance (BIAcore analysis).
  • the aforementioned antibodies can exhibit a neutral antagonist IC50 of lower than 1 x 10 -2 , or lower than 1 x 10 -3 , or 1 x 10 -4 , 1 x 10 -5 , 1 x 10 -6 , 1 x 10 -7 , 1 x 10 -8 , or 1 x 10 -9 , as determined by cellular assays.
  • the affinity and functional potency of the humanized antibody is at least comparable to the affinity and potency of a parent murine antibody.
  • the humanized antibody is not an agonist of the c-Kit receptor and does not activate mast cells which could lead to anaphylactoid reactions and should exhibit a PD/PK and immunogenicity profile that is at least comparable to the parent murine antibody.
  • a method of producing an aforementioned antibody or specific binding agent comprising culturing the aforementioned host cell such that the nucleic acid is expressed to produce the antibody or agent. Such methods may also comprise the step of recovering the antibody or agent from the host cell culture. An isolated antibody or agent produced by the aforementioned method is also disclosed.
  • the invention further provides any of the preceding antibodies or specific binding agents, for example, for use in treating or preventing a c-Kit associated disorder by administering an effective amount thereof.
  • a c-Kit associated disorder is a fibrotic disease.
  • SR-1 The murine anti-human c-Kit antibody SR-1 is described in U.S. Patent No. 5,919,911 , and U.S. Patent No. 5,489,516 .
  • SR-1 showed suitable binding properties to c-kit and blocked SCF-mediated receptor signaling, however this molecule did not possess all the characteristics that would be desired in a human therapeutic beyond the obvious immunogenicity issues. Broudy reported that SR-1 possessed some agonist-like activity that could lead to receptor internalization and phosphorylation J Cell Physiol. 1994 Mar;158(3):545-54 . These functional activities render the molecule less efficacious and less safe.
  • the humanized SR-1 in the IgG2 background proved to have high affinity for c-Kit, but in multiple cell types was unable to fully block SCF-mediated receptor internalization and in cultured mast cell assays led to c-Kit phosphorylation, a survival signal, and mediated unusual clumping of the cells. These properties are potentially undesirable since the aim of a therapeutic strategy is to apoptotically deplete mast cells and precurors by blocking the survival SCF signal and to avoid mast cell activation that could lead to anaphylactoid reactions in vivo.
  • the present inventors sought to overcome the deficiencies in each of these molecules by creating an antibody that does not have complement activation, and does not activate c-kit and mast cells while retaining desired affinity, neutral antagonist potency for the membrane c-Kit receptor, and not the soluble c-Kit receptor.
  • This antibody should also show appropriate PD/PK and is efficacious for depleting mast cells and without evidence of mast cell agonsim in vivo.
  • Seq Id No: 1 represents the nucleic acid encoding the SR-1 humanized kappa light chain.
  • Seq Id No: 2 is as follows where the bold face type represents the CDR's (e.g., CDR1 is amino acids 43 to 58 of SEQ ID NO: 2, CDR2 is amino acids 74 to 80 of SEQ ID NO: 2 and CDR3 is amino acids 113 to 121 of SEQ ID NO: 2):
  • a mature humanized kappa light chain is amino acids 20 to 248 of SEQ ID NO: 2.
  • CDR1 is amino acids 50 to 54 of SEQ ID NO: 4
  • CDR2 is amino acids 69 to 85 of SEQ ID NO: 4
  • CDR 3 is amino acids 118 to 125 of SEQ ID NO: 4):
  • the CDR's are represented in bold face type:
  • the light chain CDR1 of SR-1 is RASESVDIYGNSFMH (amino acids 44 to 58 of SEQ ID NO: 8), CDR2 is LASNLES (amino acids 74 to 80 of SEQ ID NO: 8), and CDR3 is QQNNEDPYT, (amino acids 111 to 121 of SEQ ID NO: 8).
  • Heavy chain CDR1 is SYNMH, (amino acids 50 to 54 of SEQ ID NO: 10), CDR2 is VIYSGNGDTSYNQKFKG, (amino acids 69 to 85 of SEQ ID NO: 10), CDR3 is RDTRFGN, (amino acids 118 to 125 of SEQ ID NO: 10).
  • each of the heavy and light chains depicted in the present application are processed in the cell to a mature form. Accordingly, signal peptides are cleaved and in the case of heavy chains of antibodies, the C-terminal lysine is cleaved. Accordingly, the mature form is processed proteolytically and also includes other post translational modifications such as glycosylation if expressed in mammalian cells.
  • the signal peptide for each heavy and light chain are amino acids 1 to 20 of SEQ ID NOs: 2 and 10, and amino acids 1 to 19 of SEQ ID NOs: 4, 6, and 10.
  • nucleotide and amino acid sequences of the light chain of murine SR-1 are set forth in SEQ ID NO: 7 and SEQ ID NO: 8.
  • the nucleotide and amino acid sequences of the heavy chain of murine SR-1 is set forth in SEQ ID NO: 9 and SEQ ID NO: 10.
  • Variants with further substitutions may also retain the high binding affinity. Substitutions, deletions or insertions in positions within the CDRs and framework may be made without affecting affinity.
  • the humanized antibody comprises a light chain that retains the original murine CDRs of murine SR-1, e.g ., positions about 44-58, about 74-80 and about 113-121 of SEQ ID NO: 8.
  • the humanized antibody comprises a heavy chain that retains the murine CDRs of murine SR-1, e.g ., positions about 50-54, about 68-85 and about 118-125 of SEQ ID NO:10, and has a human derived framework region.
  • the term "about” contemplates two to five amino acid position changes so long as the affinity to c-Kit is maintained.
  • Such agents comprise an amino acid sequence set forth in SEQ ID NO: 2 and an amino acid sequence that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96% , 97%, 98%, 99% or more identical to the amino acid sequence set forth in SEQ ID NO: 4; or an amino acid sequence set forth in SEQ ID NO: 4 and an amino acid sequence that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96% , 97%, 98% , 99% or more identical to the amino acid sequence set forth in SEQ ID NO: 2.
  • the sequence variation relative to SEQ ID NO: 2 or 4, respectively may represent, for example, a conservative substitution of the corresponding framework region of an IgG using an alternative human amino acid at that position.
  • the aforementioned antibodies exhibit an avidity characterized by a k d of lower than 10 -8 , or 10 -9 , as determined by surface plasmon resonance (BIAcore analysis).
  • the aforementioned antibodies exhibit a neutral antagonist potency IC50 lower than 1 x 10 -2 , or lower than 1 x 10 -3 , or 1 x 10 -4 , 1 x 10 -5 , 1 x 10 -6 , 1 x 10 -7 , 1 x 10 -8 , 1 x 10 -9 , as determined by cellular assays.
  • the present invention provides a variety of specific binding and neutral antagonist agents, including but not limited to human or humanized c-Kit specific antibodies, that are derived from murine SR-1 and retain desirable characteristics such as Kd (dissociation rate constant) for c-Kit in the range of 1 x 10 -8 or lower, or ranging down to 1 x 10 -9 or lower, and/or neutral antagonist IC50 for c-Kit in the range of 1 x 10 -2 or lower, or ranging down to 1 x 10 -9 or lower, (e.g., 10 -2 , 10 -3 , 10 -4 , 10 -5 , 10 -6 , 10 -7 , 10 -8 , 10 -9 or lower)and/or the ability to reduce symptoms of a c-Kit associated disorder.
  • Kd dissociation rate constant
  • nucleic acids encoding such specific binding agent polypeptides, vectors and recombinant host cells comprising such nucleic acids, methods of producing such specific binding agents, pharmaceutical formulations including such specific binding agents, methods of preparing the pharmaceutical formulations, and methods of treating patients with the pharmaceutical formulations and compounds.
  • Nucleic acids encoding these modified light chain variable regions were constructed and co-expressed with nucleic acids encoding a CDR-grafted or a humanized heavy chain and vice versa, and optionally may be linked to constant regions. Any humanized or chimeric heavy chain and light chains may be combined as long as suitable binding affinity is maintained.
  • the desired genes were introduced into mammalian cells and the resultant recombinant immunoglobulin products were expressed, purified and characterized.
  • antibody is used in the broadest sense and includes fully assembled antibodies, monoclonal antibodies (including human, humanized or chimeric antibodies), polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies), and antibody fragments that can bind antigen (e.g., Fab', F'(ab)2, Fv, single chain antibodies, diabodies), comprising complementarity determining regions (CDRs) of the foregoing as long as they exhibit the desired biological activity.
  • CDRs complementarity determining regions
  • the term “antibody” explicitly excludes murine antibody (i.e. antibody produced by a murine hybridoma or having the same sequence as an antibody produced by a murine hybridoma) from the scope of the term.
  • specific binding agent includes antibodies as defined above and recombinant peptides or other compounds that contain sequences derived from CDRs having the desired antigen-binding properties. Specifically included in the term are peptides containing amino acid sequences that are at least 80%, 90% or 100% identical to one or more CDRs of murine SR-1, preferably including heavy chain CDR3.
  • neutral antagonist is understood to mean a specific binding agent that is capable of inhibiting an agonists activity
  • agents includes antibodies as defined above and recombinant peptides or other compounds that contain sequences derived from CDRs having the desired antigen-binding properties.
  • peptides containing amino acid sequences that are at least 80%, 90% or 100% identical to one or more CDRs of murine SR-1, preferably including heavy chain CDR3.
  • Peptibodies which are molecules comprising an antibody Fc domain as the "vehicle” attached to at least one antigen-binding peptide.
  • Antibody CDR's from the SR-1 antibody may be suitable for incorporation into a peptibody, particularly including the CDR3 of the heavy chain.
  • the production of peptibodies is generally described in PCT publication WO 00/24782, published May 4, 2000 .
  • Peptides may be linked in tandem (i.e., sequentially), with or without linkers.
  • Peptides containing a cysteinyl residue may be cross-linked with another Cys-containing peptide, either or both of which may be linked to a vehicle.
  • Any peptide having more than one Cys residue may form an intrapeptide disulfide bond, as well. Any of these peptides may be derivatized, for example the carboxyl terminus may be capped with an amino group, cysteines may be capped, or amino acid residues may substituted by moieties other than amino acid residues (see, e.g., Bhatnagar et al., J. Med. Chem. 39: 3814-9 (1996 ), and Cuthbertson et al., J. Med. Chem. 40: 2876-82 (1997 )).
  • the peptide sequences may be optimized, analogous to affinity maturation for antibodies, or otherwise altered by alanine scanning or random or directed mutagenesis followed by screening to identify the best binders. Lowman, Ann. Rev. Biophys. Biomol. Struct. 26: 401-24 (1997 ).
  • Various molecules can be inserted into the specific binding agent structure, e.g., within the peptide portion itself or between the peptide and vehicle portions of the specific binding agents, while retaining the desired activity of specific binding agent.
  • molecules such as an Fc domain or fragment thereof, polyethylene glycol or other related molecules such as dextran, a fatty acid, a lipid, a cholesterol group, a small carbohydrate, a peptide, a cyotoxic agent, a chemotherapeutic agent, a detectable moiety as described herein (including fluorescent agents, radiolabels such as radioisotopes), an oligosaccharide, oligonucleotide, a polynucleotide, interference (or other) RNA, enzymes, hormones, or the like.
  • Other molecules suitable for insertion in this fashion will be appreciated by those skilled in the art, and are encompassed within the scope of the invention. This includes insertion of, for example, a desired molecule in between two
  • an "isolated” antibody is one that has been identified and separated from a component of the cell that expressed it. Contaminant components of the cell are materials that would interfere with diagnostic or therapeutic uses for the antibody, and may include enzymes, hormones, and other proteinaceous or nonproteinaceous solutes.
  • the antibody will be purified (1) to greater than 95% by weight of antibody, and most preferably more than 99% by weight, (2) to a degree sufficient to obtain at least 15 residues of N-terminal or internal amino acid sequence, or (3) to homogeneity by SDS-PAGE under reducing or nonreducing conditions using Coomassie blue or, preferably, silver stain.
  • Isolated naturally occurring antibody includes the antibody in situ within recombinant cells since at least one component of the antibody's natural environment will not be present. Ordinarily, however, isolated antibody will be prepared by at least one purification step.
  • monoclonal antibody refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against an individual antigenic site or epitope, in contrast to polyclonal antibody preparations that typically include different antibodies directed against different epitopes.
  • human immunoglobulins can be assigned to different classes. There are five major classes, IgA, IgD, IgE, IgG and IgM, and several of these may be further divided into subclasses or isotypes, e.g. IgG1, IgG2, IgG3, IgG4, IgAl and IgA2.
  • the heavy-chain constant domains that correspond to the different classes of immunoglobulins are called alpha, delta, epsilon, gamma and mu respectively.
  • the subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known. Different isotypes have different effector functions; for example, IgG1 and IgG3 isotypes have ADCC activity.
  • hypervariable region refers to the amino acid residues of an antibody which are responsible for antigen-binding.
  • the hypervariable region comprises amino acid residues from a complementarity determining region or CDR [i.e., residues 24-34 (L1), 50-56 (L2) and 89-97 (L3) in the light chain variable domain and 31-35 (HI), 50-65 (H2) and 95-102 (H3) in the heavy chain variable domain as described by Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991 )].
  • CDR Even a single CDR may recognize and bind antigen, although with a lower affinity than the entire antigen binding site containing all of the CDRs. It is understood that the CDR of an antibody may include additional or fewer sequences outside the specified limits above so long as the antibody retains it's ability to bind the target molecule.
  • residues from a hypervariable "loop” is described by Chothia et al., J. Mol.Biol. 196: 901-917 (1987 ) as residues 26-32 (LI), 50-52 (L2) and 91-96 (L3) in the light chain variable domain and 26-32 (HI), 53-55 (H2) and 96-101 (H3) in the heavy chain variable domain .
  • Framework or FR residues are those variable region residues other than the hypervariable region residues.
  • Antibody fragments comprise a portion of an intact full length antibody, preferably the antigen binding or variable region of the intact antibody.
  • antibody fragments include Fab, Fab', F(ab')2, and Fv fragments; diabodies; linear antibodies ( Zapata et al., Protein Eng.,8(10):1057-1062 (1995 )); single-chain antibody molecules; and multispecific antibodies formed from antibody fragments.
  • Papain digestion of antibodies produces two identical antigen-binding fragments, called “Fab” fragments, each with a single antigen-binding site, and a residual "Fc” fragment which contains the constant region.
  • the Fab fragment contains all of the variable domain, as well as the constant domain of the light chain and the first constant domain (C H 1) of the heavy chain.
  • the Fc fragment displays carbohydrates and is responsible for many antibody effector functions (such as binding complement and cell receptors), that distinguish one class of antibody from another.
  • Fv polypeptide further comprises a polypeptide linker between the V H and V L domains that enables the Fv to form the desired structure for antigen binding.
  • Fv is the minimum antibody fragment that contains a complete antigen recognition and binding site. This region consists of a dimer of one heavy- and one light-chain variable domain in tight, non-covalent association. It is in this configuration that the three CDRs of each variable domain interact to define an antigen binding site on the surface of the V H V L dimer. Collectively, the six CDRs confer antigen-binding specificity to the antibody. However, a single variable domain (or half of an Fv comprising only three CDRs specific for an antigen) has the ability to recognize and bind antigen, although at a lower affinity than the entire binding site.
  • diabodies refers to small antibody fragments with two antigen-binding sites, which fragments comprise a heavy-chain variable domain (V H ) connected to a light-chain variable domain (V L ) in the same polypeptide chain (V H V L ).
  • V H heavy-chain variable domain
  • V L light-chain variable domain
  • the domains are forced to pair with the complementary domains of another chain and create two antigen-binding sites.
  • Diabodies are described more fully in, for example, EP 404,097 ; WO 93/11161 ; and 30 Hollinger et al., Proc. Natl. Acad. Sci. USA, 90:6444-6448 (1993 ).
  • compositions and methods of treating inflammatory, autoimmune, oncologic and fibrotic disorders may utilize one or more anti-c-Kit therapeutics used singularly or in combination with other therapeutics to achieve the desired effects.
  • exemplary anti-fibrotic agents suitable for use in accordance with the invention include cytokines wherein the cytokine is selected from transforming growth factor ⁇ (TGF- ⁇ ), interleukin- 4 (IL-4), interleukin-5 (IL-5), interleukin-9 (IL-9), interleukin-13(IL-13), granulocyte/macrophage-colony stimulating factor (GM-CSF), tumor necrosis factor alpha (TNF- ⁇ ), interleukin-1 beta (IL-1 ⁇ ), connective tissue growth factor (CTGF), interleukin-6 (IL-6), oncostatin M (OSM), platelet derived growth factor (PDGF), monocyte chemotactic protein 1 (CCL2/MCP-1), and pulmonary and activation- regulated chemokine (CCL18/PARC
  • Antibodies derived from SR-1 according to the present invention are preferably produced by recombinant DNA methodology using one of the antibody expression systems well known in the art (see, e.g., Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory (1988 )). Such antibodies are also preferably chimeric fusion proteins having immunoglobulin derived variable sequences and human constant regions, or more preferably are more human-like monoclonal antibodies (such as human or humanized antibodies) that comprise human antibody residues but preferably retain at least the CDRs of murine SR-1.
  • the term "antibody” also refers to fragments thereof or multimers or aggregates of intact molecules and/or fragments that bind to c-Kit.
  • humanized antibody refers to an antibody derived from a non-human antibody, typically a mouse monoclonal antibody.
  • a humanized antibody may be derived from a chimeric antibody that retains or substantially retains the antigen binding properties of the parental, non-human, antibody but which exhibits diminished immunogenicity as compared to the parental antibody when administered to humans.
  • chimeric antibody refers to an antibody containing sequence derived from two different antibodies (see, e.g., U.S. Patent No. 4,816,567 ) which typically originate from different species. Most typically, chimeric antibodies comprise human and murine antibody fragments, generally human constant and mouse variable regions.
  • amino acid sequence of an immunoglobulin of interest may be determined by direct protein sequencing, and suitable encoding nucleotide sequences can be designed according to a universal codon table.
  • DNA encoding the monoclonal antibodies may be isolated and sequenced from the hybridoma cells using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of the monoclonal antibodies). Sequence determination will generally require isolation of at least a portion of the gene or cDNA of interest. Usually this requires cloning the DNA or, preferably, mRNA (i.e., cDNA) encoding the monoclonal antibodies. Cloning is carried out using standard techniques (see, e.g., Sambrook et al. (1989) Molecular Cloning: A Laboratory Guide, Vols 1-3, Cold Spring Harbor Press ).
  • a cDNA library may be constructed by reverse transcription of polyA+ mRNA, preferably membrane-associated mRNA, and the library screened using probes specific for human immunoglobulin polypeptide gene sequences.
  • the polymerase chain reaction PCR
  • PCR polymerase chain reaction
  • the amplified sequences can be readily cloned into any suitable vector, e.g., expression vectors, minigene vectors, or phage display vectors. It will be appreciated that the particular method of cloning used is not critical, so long as it is possible to determine the sequence of some portion of the immunoglobulin polypeptide of interest.
  • an "isolated" nucleic acid molecule or “isolated” nucleic acid sequence is a nucleic acid molecule that is either (1) identified and separated from at least one contaminant nucleic acid molecule with which it is ordinarily associated in the natural source of the nucleic acid or (2) cloned, amplified, tagged, or otherwise distinguished from background nucleic acids such that the sequence of the nucleic acid of interest can be determined.
  • An isolated nucleic acid molecule is other than in the form or setting in which it is found in nature.
  • an isolated nucleic acid molecule includes a nucleic acid molecule contained in cells that ordinarily express the antibody where, for example, the nucleic acid molecule is in a chromosomal location different from that of natural cells.
  • RNA used for cloning and sequencing is a hybridoma produced by obtaining a B cell from the transgenic mouse and fusing the B cell to an immortal cell.
  • An advantage of using hybridomas is that they can be easily screened, and a hybridoma that produces a human monoclonal antibody of interest selected.
  • RNA can be isolated from B cells (or whole spleen) of the immunized animal.
  • sources other than hybridomas it may be desirable to screen for sequences encoding immunoglobulins or immunoglobulin polypeptides with specific binding characteristics.
  • One method for such screening is the use of phage display technology.
  • Phage display is described in e.g., Dower et al., WO 91/17271 , McCafferty et al., WO 92/01047 , and Caton and Koprowski, Proc. Natl. Acad. Sci. USA, 87:6450-6454 (1990 ).
  • cDNA from an immunized transgenic mouse e.g., total spleen cDNA
  • the polymerase chain reaction is used to amplify a cDNA sequences that encode a portion of an immunoglobulin polypeptide, e.g., CDR regions, and the amplified sequences are inserted into a phage vector.
  • cDNAs encoding peptides of interest e.g., variable region peptides with desired binding characteristics, are identified by standard techniques such as panning.
  • sequence of the amplified or cloned nucleic acid is then determined.
  • sequence encoding an entire variable region of the immunoglobulin polypeptide is determined, however, it will sometimes be adequate to sequence only a portion of a variable region, for example, the CDR-encoding portion.
  • portion sequenced will be at least 30 bases in length, more often bases coding for at least about one-third or at least about one-half of the length of the variable region will be sequenced.
  • Sequencing can be carried out on clones isolated from a cDNA library, or, when PCR is used, after subcloning the amplified sequence or by direct PCR sequencing of the amplified segment. Sequencing is carried out using standard techniques (see, e.g., Sambrook et al. (1989) Molecular Cloning: A Laboratory Guide, Vols 1-3, Cold Spring Harbor Press , and Sanger, F. et al. (1977) Proc. Natl. Acad. Sci. USA 74: 5463-5467 ).
  • the DNA may be operably linked to expression control sequences or placed into expression vectors, which are then transfected into host cells such as E. coli cells, simian COS cells, Chinese hamster ovary (CHO) cells, or myeloma cells that do not otherwise produce immunoglobulin protein, to direct the synthesis of monoclonal antibodies in the recombinant host cells. Recombinant production of antibodies is well known in the art.
  • Expression control sequences refer to DNA sequences necessary for the expression of an operably linked coding sequence in a particular host organism.
  • the control sequences that are suitable for prokaryotes include a promoter, optionally an operator sequence, and a ribosome binding site.
  • Eukaryotic cells are known to utilize promoters, polyadenylation signals, and enhancers.
  • Nucleic acid is operably linked when it is placed into a functional relationship with another nucleic acid sequence.
  • DNA for a presequence or secretory leader is operably linked to DNA for a polypeptide if it is expressed as a preprotein that participates in the secretion of the polypeptide;
  • a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence; or
  • a ribosome binding site is operably linked to a coding sequence if it is positioned so as to facilitate translation.
  • operably linked means that the DNA sequences being linked are contiguous, and, in the case of a secretory leader, contiguous and in reading phase. However, enhancers do not have to be contiguous. Linking is accomplished by ligation at convenient restriction sites. If such sites do not exist, the synthetic oligonucleotide adaptors or linkers are used in accordance with conventional practice.
  • progeny include the primary subject cell and cultures derived therefrom without regard for the number of transfers. It is also understood that all progeny may not be precisely identical in DNA content, due to deliberate or inadvertent mutations. Mutant progeny that have the same function or biological activity as screened for in the originally transformed cell are included.
  • the invention also provides isolated nucleic acids encoding specific binding agents or antibodies of the invention, optionally operably linked to control sequences recognized by a host cell, vectors and host cells comprising the nucleic acids, and recombinant techniques for the production of the specific binding agents or antibodies, which may comprise culturing the host cell so that the nucleic acid is expressed and, optionally, recovering the specific binding agent or antibody from the host cell culture or culture medium.
  • Vector components may include one or more of the following: a signal sequence (that may, for example, direct secretion of the specific binding agent or antibody), an origin of replication, one or more selective marker genes (that may, for example, confer antibiotic or other drug resistance, complement auxotrophic deficiencies, or supply critical nutrients not available in the media), an enhancer element, a promoter, and a transcription termination sequence, all of which are well known in the art.
  • Suitable host cells include prokaryote, yeast, or higher eukaryote cells described above.
  • Suitable prokaryotes for this purpose include eubacteria, such as Gram-negative or Gram-positive organisms, for example, Enterobacteriaceae such as Escherichia, e.g., E. coli, Enterobacter, Erwinia, Klebsiella, Proteus, Salmonella, e.g., Salmonella typhimurium, Serratia, e.g., Serratia marcescans, and Shigella, as well as Bacilli such as B. subtilis and B. licheniformis, Pseudomonas, and Streptomyces.
  • Enterobacteriaceae such as Escherichia, e.g., E. coli, Enterobacter, Erwinia, Klebsiella, Proteus
  • Salmonella e.g., Salmonella typhimurium
  • Serratia
  • eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for specific binding agent-encoding vectors.
  • Saccharomyces cerevisiae or common baker's yeast, is the most commonly used among lower eukaryotic host microorganisms.
  • a number of other genera, species, and strains are commonly available and useful herein, such as Pichia, e.g. P. pastoris, Schizosaccharomyces pombe; Kluyveromyces, Yarrowia; Candida; Trichoderma reesia;
  • Neurospora crassa Schwanniomyces such as Schwanniomyces occidentalis
  • filamentous fungi such as, e.g., Neurospora, Penicillium, Tolypocladium, and Aspergillus hosts such as A. nidulans and A. niger.
  • Suitable host cells for the expression of glycosylated specific binding agent or antibody are derived from multicellular organisms.
  • invertebrate cells include plant and insect cells.
  • Numerous baculoviral strains and variants and corresponding permissive insect host cells from hosts such as Spodoptera frugiperda (caterpillar), Aedes aegypti (mosquito), Aedes albopictus (mosquito), Drosophila melanogaster (fruitfly), and Bombyx mori have been identified.
  • a variety of viral strains for transfection of such cells are publicly available, e.g., the L-1 variant of Autographa californica NPV and the Bm-5 strain of Bombyx mori NPV.
  • ⁇ hamster ovary cells including CHOK1 cells (ATCC CCL61), DXB-11, DG-44, and Chinese hamster ovary cells/-DHFR ( CHO, Urlaub et al., Proc. Natl. Acad. Sci. USA 77: 4216 (1980 )); monkey kidney CV1 line transformed by SV40 (COS-7, ATCC CRL 1651); human embryonic kidney line (293 or 293 cells subcloned for growth in suspension culture, [ Graham et al., J. Gen Virol.
  • Host cells are transformed or transfected with the above-described nucleic acids or vectors for specific binding agent or antibody production and cultured in conventional nutrient media modified as appropriate for inducing promoters, selecting transformants, or amplifying the genes encoding the desired sequences.
  • novel vectors and transfected cell lines with multiple copies of transcription units separated by a selective marker are particularly useful and preferred for the expression of specific binding agents or antibodies.
  • the host cells used to produce the specific binding agent or antibody of this invention may be cultured in a variety of media.
  • Commercially available media such as Ham's F10 (Sigma), Minimal Essential Medium ((MEM), (Sigma), RPMI-1640 (Sigma), and Dulbecco's Modified Eagle's Medium ((DMEM), Sigma) are suitable for culturing the host cells.
  • any of these media may be supplemented as necessary with hormones and/or other growth factors (such as insulin, transferrin, or epidermal growth factor), salts (such as sodium chloride, calcium, magnesium, and phosphate), buffers (such as HEPES), nucleotides (such as adenosine and thymidine), antibiotics (such as GentamycinTM drug), trace elements (defined as inorganic compounds usually present at final concentrations in the micromolar range), and glucose or an equivalent energy source. Any other necessary supplements may also be included at appropriate concentrations that would be known to those skilled in the art.
  • the culture conditions such as temperature, pH, and the like, are those previously used with the host cell selected for expression, and will be apparent to the ordinarily skilled artisan.
  • the expression vectors, pDC323 and pDC324 as described in U.S. Patent Application No. 20030082735 containing the appropriate respective light chain and heavy chain pair were transfected into the CS9 host cell line.
  • the specific binding agent or antibody can be produced intracellularly, in the periplasmic space, or directly secreted into the medium. If the specific binding agent or antibody is produced intracellularly, as a first step, the particulate debris, either host cells or lysed fragments, is removed, for example, by centrifugation or ultrafiltration.
  • the specific binding agent or antibody composition can be purified using, for example, hydroxylapatite chromatography, cation or anion exchange chromatography, or preferably affinity chromatography, using the antigen of interest or protein A or protein G as an affinity ligand.
  • Protein A can be used to purify specific binding agents or antibodies that are based on human ⁇ 1, ⁇ 2, or ⁇ 4 heavy chains ( Lindmark et al., J. Immunol. Meth. 62: 1-13 (1983 )).
  • Protein G is recommended for all mouse isotypes and for human ⁇ 3 ( Guss et al., EMBO J. 5: 15671575 (1986 )).
  • the matrix to which the affinity ligand is attached is most often agarose, but other matrices are available.
  • Mechanically stable matrices such as controlled pore glass or poly(styrenedivinyl)benzene allow for faster flow rates and shorter processing times than can be achieved with agarose.
  • the specific binding agent or antibody comprises a C H 3 domain
  • the Bakerbond ABXTMresin J. T. Baker, Phillipsburg, N.J.
  • Other techniques for protein purification such as ethanol precipitation, Reverse Phase HPLC, chromatofocusing, SDS-PAGE, and ammonium sulfate precipitation are also possible depending on the specific binding agent or antibody to be recovered.
  • Chimeric monoclonal antibodies in which the variable Ig domains of a rodent monoclonal antibody are fused to human constant Ig domains, can be generated using standard procedures known in the art (See Morrison, S. L., et al. (1984) Chimeric Human Antibody Molecules; Mouse Antigen Binding Domains with Human Constant Region Domains, Proc. Natl. Acad. Sci. USA 81, 6841-6855 ; and, Boulianne, G. L., et al, Nature 312, 643-646 . (1984 )). Although some chimeric monoclonal antibodies have proved less immunogenic in humans, the rodent variable Ig domains can still lead to a significant human anti-rodent response.
  • Humanized antibodies may be achieved by a variety of methods including, for example: (1) grafting the non-human complementarity determining regions (CDRs) onto a human framework and constant region (a process referred to in the art as humanizing through “CDR grafting"), or, alternatively, (2) transplanting the entire non-human variable domains, but “cloaking” them with a human-like surface by replacement of surface residues (a process referred to in the art as "veneering”).
  • CDRs complementarity determining regions
  • a rodent antibody on repeated in vivo administration in man either alone or as a conjugate will bring about an immune response in the recipient against the rodent antibody; the so-called HAMA response (Human Anti Mouse Antibody).
  • HAMA response Human Anti Mouse Antibody
  • the HAMA response may limit the effectiveness of the pharmaceutical if repeated dosing is required.
  • the immunogenicity of the antibody may be reduced by chemical modification of the antibody with a hydrophilic polymer such as polyethylene glycol or by using the methods of genetic engineering to make the antibody binding structure more human like.
  • CDR grafting involves introducing one or more of the six CDRs from the mouse heavy and light chain variable Ig domains into the appropriate framework regions of a human variable Ig domain.
  • This technique Riechmann, L., et al., Nature 332, 323 (1988 )
  • FR1-FR4 conserved framework regions
  • a significant disadvantage of CDR grafting is that it can result in a humanized antibody that has a substantially lower binding affinity than the original mouse antibody, because amino acids of the framework regions can contribute to antigen binding, and because amino acids of the CDR loops can influence the association of the two variable Ig domains.
  • Chimeric SR-1 antibodies did not show appropriate functional potency in cell based assays.
  • the CDR grafting technique can be improved by choosing human framework regions that most closely resemble the framework regions of the original mouse antibody, and by site-directed mutagenesis of single amino acids within the framework or CDRs aided by computer modeling of the antigen binding site (e.g., Co, M. S., et al. (1994), J. Immunol. 152, 2968-2976 ).
  • One method of humanizing antibodies comprises aligning the non-human heavy and light chain sequences to human heavy and light chain sequences, selecting and replacing the non-human framework with a human framework based on such alignment, molecular modeling to predict the conformation of the humanized sequence and comparing to the conformation of the parent antibody. This process is followed by repeated back mutation of residues in the CDR region which disturb the structure of the CDRs until the predicted conformation of the humanized sequence model closely approximates the conformation of the non-human CDRs of the parent non-human antibody.
  • Such humanized antibodies may be further derivatized to facilitate uptake and clearance, e.g., via Ashwell receptors (See, e.g., U.S. Patent Nos. 5,530,101 and 5,585,089 ).
  • Amino acid sequence variants of the desired specific binding agent or antibody may be prepared by introducing appropriate nucleotide changes into the encoding DNA, or by peptide synthesis. Such variants include, for example, deletions and/or insertions and/or substitutions of residues within the amino acid sequences of the specific binding agents or antibodies. Any combination of deletion, insertion, and substitution is made to arrive at the final construct, provided that the final construct possesses the desired characteristics.
  • the amino acid changes also may alter post-translational processes of the specific binding agent or humanized or variant antibody, such as changing the number or position of glycosylation sites.
  • Nucleic acid molecules encoding amino acid sequence variants of the specific binding agent or antibody are prepared by a variety of methods known in the art. Such methods include oligonucleotide-mediated (or site-directed) mutagenesis, PCR mutagenesis, and cassette mutagenesis of an earlier prepared variant or a non-variant version of the specific binding agent or antibody.
  • alanine scanning mutagenesis A useful method for identification of certain residues or regions of the specific binding agent or antibody that are preferred locations for mutagenesis is called " alanine scanning mutagenesis," as described by Cunningham and Wells Science, 244:1081-1085 (1989 ).
  • a residue or group of target residues are identified (e.g., charged residues such as arg, asp, his, lys, and glu) and replaced by a neutral or negatively charged amino acid (most preferably alanine or polyalanine) to affect the interaction of the amino acids with antigen.
  • Those amino acid locations demonstrating functional sensitivity to the substitutions then are refined by introducing further or other variants at, or for, the sites of substitution.
  • the site for introducing an amino acid sequence variation is predetermined, the nature of the mutation per se need not be predetermined. For example, to analyze the performance of a mutation at a given site, ala scanning or random mutagenesis is conducted at the target codon or region and the expressed variants are screened for the desired activity.
  • amino acid sequence variants of the specific antibody will have an amino acid sequence having at least 90% amino acid sequence identity with the original specific binding agent or antibody (murine or humanized) amino acid sequences of either the heavy or the light chain, or at least 95% identity, including for example, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, and 100%.
  • Identity or homology with respect to this sequence is defined herein as the percentage of amino acid residues in the candidate sequence that are identical with the original sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions (as defined in Table I below) as part of the sequence identity.
  • sequence identity can be determined by standard methods that are commonly used to compare the similarity in position of the amino acids of two polypeptides. Using a computer program such as BLAST or FASTA, two polypeptides are aligned for optimal matching of their respective amino acids (either along the full length of one or both sequences, or along a pre-determined portion of one or both sequences). The programs provide a default opening penalty and a default gap penalty, and a scoring matrix such as PAM 250 [a standard scoring matrix; see Dayhoff et al., in Atlas of Protein Sequence and Structure, vol.
  • the percent identity can then be calculated as: the total number of identical matches multiplied by 100 and then divided by the sum of the length of the longer sequence within the matched span and the number of gaps introduced into the longer sequences in order to align the two sequences.
  • Amino acid sequence insertions include amino- and/or carboxyl-terminal fusions ranging in length from one residue to polypeptides containing a hundred or more residues, as well as intra-sequence insertions of single or multiple amino acid residues.
  • terminal insertions include a specific binding agent or antibody with an N-terminal methionyl residue or the specific binding agent or antibody (including antibody fragment) fused to an epitope tag or a salvage receptor epitope.
  • Other insertional variants of the specific binding agent or antibody molecule include the fusion to a polypeptide which increases the serum half-life of the specific binding agent or antibody, e.g. at the N-terminus or C-terminus.
  • epitope tags include the flu HA tag polypeptide and its antibody 12CA5 [ Field et al., Mol. Cell. Biol. 8: 2159-2165 (1988 )]; the c-myc tag and the 8F9, 3C7, 6E10, G4, B7 and 9E10 antibodies thereto [ Evan et al., Mol. Cell. Biol. 5(12): 3610-3616 (1985 )]; and the Herpes Simplex virus glycoprotein D (gD) tag and its antibody [ Paborsky et al., Protein Engineering 3(6): 547-553 (1990 )].
  • tags are a poly-histidine sequence, generally around six histidine residues, that permits isolation of a compound so labeled using nickel chelation.
  • Other labels and tags such as the FLAG® tag (Eastman Kodak, Rochester, NY) are well known and routinely used in the art.
  • the term "salvage receptor binding epitope” refers to an epitope of the Fc region of an IgG molecule (e.g., IgG 1 , IgG 2 , IgG 3 , or IgG 4 ) that is responsible for increasing the in vivo serum half-life of the IgG molecule.
  • variants are an amino acid substitution variant. These variants have at least one amino acid residue in the specific binding agent or antibody molecule removed and a different residue inserted in its place. Substitutional mutagenesis within any of the hypervariable or CDR regions or framework regions is contemplated. Conservative substitutions are shown in Table 1. The most conservative substitution is found under the heading of "preferred substitutions”. If such substitutions result in no change in biological activity, then more substantial changes, denominated "exemplary substitutions" in Table 1, or as further described below in reference to amino acid classes, may be introduced and the products screened.
  • Substantial modifications in the biological properties of the specific binding agent or antibody are accomplished by selecting substitutions that differ significantly in their effect on maintaining (a) the structure of the polypeptide backbone in the area of the substitution, for example, as a sheet or helical conformation, (b) the charge or hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain.
  • Naturally occurring residues are divided into groups based on common side-chain properties:
  • Conservative substitutions involve replacing an amino acid with another member of its class.
  • Non-conservative substitutions involve replacing a member of one of these classes with a member of another class.
  • cysteine residues not involved in maintaining the proper conformation of the specific binding agent or humanized or variant antibody also may be substituted, generally with serine, to improve the oxidative stability of the molecule and prevent aberrant crosslinking.
  • cysteine bond(s) may be added to the specific binding agent or antibody to improve its stability (particularly where the antibody is an antibody fragment such as an Fv fragment).
  • Affinity maturation involves preparing and screening specific binding agent or antibody variants that have mutations (deletions, insertions or substitutions) within the CDRs of a parent specific binding agent or antibody and selecting variants that have improved biological properties such as binding affinity relative to the parent specific binding agent or antibody.
  • a convenient way for generating such substitutional variants is affinity maturation using phage display. Briefly, several hypervariable region sites (e.g. 6-7 sites) are mutated to generate all possible amino substitutions at each site.
  • the specific binding agent or antibody variants thus generated may be displayed in a monovalent fashion from filamentous phage particles as fusions to the gene III product of M13 packaged within each particle. The phage-displayed variants are then screened for their biological activity (e.g. binding affinity).
  • Alanine scanning mutagenesis can be performed to identify hypervariable region residues that contribute significantly to antigen binding. Alternatively, or in addition, it may be beneficial to analyze a crystal structure of the antigen-antibody complex to identify contact points between the specific binding agent or antibody and the antigen. Such contact residues and neighboring residues are candidates for substitution according to the techniques elaborated herein. Once such variants are generated, the panel of variants is subjected to screening as described herein and specific binding agents or antibodies with superior properties in one or more relevant assays may be selected for further development.
  • Specific binding agent or antibody variants can also be produced that have a modified glycosylation pattern relative to the parent specific binding agent or antibody, for example, deleting one or more carbohydrate moieties found in the specific binding agent or antibody, and/or adding one or more glycosylation sites that are not present in the specific binding agent or antibody.
  • N-linked refers to the attachment of the carbohydrate moiety to the side chain of an asparagine residue.
  • the tripeptide sequences asparagine-X-serine and asparagine-X-threonine, where X is any amino acid except proline, are the recognition sequences for enzymatic attachment of the carbohydrate moiety to the asparagine side chain.
  • the presence of either of these tripeptide sequences in a polypeptide creates a potential glycosylation site.
  • N-linked glycosylation sites may be added to a specific binding agent or antibody by altering the amino acid sequence such that it contains one or more of these tripeptide sequences.
  • O-linked glycosylation refers to the attachment of one of the sugars N-aceylgalactosamine, galactose, or xylose to a hydroxyamino acid, most commonly serine or threonine, although 5-hydroxyproline or 5-hydroxylysine may also be used.
  • O-linked glycosylation sites may be added to a specific binding agent or antibody by inserting or substituting one or more serine or threonine residues to the sequence of the original specific binding agent or antibody.
  • Cysteine residue(s) may be removed or introduced in the Fc region, thereby eliminating or increasing interchain disulfide bond formation in this region.
  • the homodimeric specific binding agent or antibody thus generated may have improved internalization capability and/or increased complement-mediated cell killing and antibody-dependent cellular cytotoxicity (ADCC).
  • ADCC antibody-dependent cellular cytotoxicity
  • Homodimeric specific binding agents or antibodies may also be prepared using heterobifunctional cross-linkers as described in Wolff et al., Cancer Research 53: 2560-2565 (1993 ).
  • a specific binding agent or antibody can be engineered which has dual Fc regions and may thereby have enhanced complement lysis and ADCC capabilities. See Stevenson et al., Anti-CancerDrug Design 3: 219-230 (1989 ).
  • sequences within the CDR can cause an antibody to bind to MHC Class II and trigger an unwanted helper T-cell response.
  • a conservative substitution can allow the specific binding agent or antibody to retain binding activity yet reduce its ability to trigger an unwanted T-cell response.
  • N-terminal 20 amino acids of the heavy or light chain are removed.
  • Modifications to increase serum half-life also may desirable, for example, by incorporation of or addition of a salvage receptor binding epitope (e.g., by mutation of the appropriate region or by incorporating the epitope into a peptide tag that is then fused to the specific binding agent or antibody at either end or in the middle, e.g., by DNA or peptide synthesis) (see, e.g., WO96/32478 ) or adding molecules such as PEG or other water soluble polymers, including polysaccharide polymers.
  • a salvage receptor binding epitope e.g., by mutation of the appropriate region or by incorporating the epitope into a peptide tag that is then fused to the specific binding agent or antibody at either end or in the middle, e.g., by DNA or peptide synthesis
  • molecules such as PEG or other water soluble polymers, including polysaccharide polymers.
  • the salvage receptor binding epitope preferably constitutes a region wherein any one or more amino acid residues from one or two loops of a Fc domain are transferred to an analogous position of the specific binding agent or antibody or fragment. Even more preferably, three or more residues from one or two loops of the Fc domain are transferred. Still more preferred, the epitope is taken from the C H 2 domain of the Fc region (e.g., of an IgG) and transferred to the C H 1, C H 3, or V H region, or more than one such region, of the specific binding agent or antibody. Alternatively, the epitope is taken from the C H 2 domain of the Fc region and transferred to the C L region or V L region, or both, of the specific binding agent or antibody fragment. See also International applications WO 97/34631 and WO 96/32478 which describe Fc variants and their interaction with the salvage receptor.
  • IgG homeostasis in vivo depends upon its binding to the FcRn. Modification of the interaction between the Fc domain of IgG and FcRn has been reported to improve the serum half-life of monoclonal antibodies. Mutations in the Fc that result in higher affinity binding to neonatal Fc receptor FcRn and slowing degradation and improving PK profile would be preferable.
  • the FcRn binding site on IgG is located at the C H 2-C H 3 domain interface.
  • Mutations of residues in this area result in increased affinity of IgG1 for human FcRn at pH 6.0 and pH 7.3. Additionally, some of these mutations resulted in enhanced pharmacokinetic properties (slower clearance, longer half-life) when given intravenously to monkeys.
  • CDC complement dependent cytotoxicity
  • ADCC antibody-dependent cellular cytotoxicity
  • potential mutations include insertion, deletion or substitution of one or more residues, including substitution with alanine, a conservative substitution, a non-conservative substitution, or replacement with a corresponding amino acid residue at the same position from a different subclass (e.g. replacing an IgG1 residue with a corresponding IgG2 residue at that position).
  • Covalent modifications of the specific binding agent or antibody are also included within the scope of this invention. They may be made by chemical synthesis or by enzymatic or chemical cleavage of the specific binding agent or antibody, if applicable. Other types of covalent modifications can be introduced into the specific binding agent or antibody by reacting targeted amino acid residues with an organic derivatizing agent that is capable of reacting with selected side chains or the N- or C-terminal residues.
  • Cysteinyl residues most commonly are reacted with ⁇ -haloacetates (and corresponding amines), such as chloroacetic acid or chloroacetamide, to give carboxymethyl or carboxyamidomethyl derivatives. Cysteinyl residues also are derivatized by reaction with bromotrifluoroacetone, .alpha.-bromo- ⁇ -(5-imidozoyl)propionic acid, chloroacetyl phosphate, N-alkylmaleimides, 3-nitro-2-pyridyl disulfide, methyl 2-pyridyl disulfide, p-chloromercuribenzoate, 2-chloromercuri-4-nitrophenol, or chloro-7-nitrobenzo-2-oxa-1,3-diazole.
  • Histidyl residues are derivatized by reaction with diethylpyrocarbonate at pH 5.5-7.0 because this agent is relatively specific for the histidyl side chain.
  • Para-bromophenacyl bromide also is useful; the reaction is preferably performed in 0.1 M sodium cacodylate at pH 6.0.
  • Lysinyl and amino-terminal residues are reacted with succinic or other carboxylic acid anhydrides. Derivatization with these agents has the effect of reversing the charge of the lysinyl residues.
  • Other suitable reagents for derivatizing .alpha.-amino-containing residues include imidoesters such as methyl picolinimidate, pyridoxal phosphate, pyridoxal, chloroborohydride, trinitrobenzenesulfonic acid, O-methylisourea, 2,4-pentanedione, and transaminase-catalyzed reaction with glyoxylate.
  • Arginyl residues are modified by reaction with one or several conventional reagents, among them phenylglyoxal, 2,3-butanedione, 1,2-cyclohexanedione, and ninhydrin. Derivatization of arginine residues requires that the reaction be performed in alkaline conditions because of the high pK a of the guanidine functional group. Furthermore, these reagents may react with the groups of lysine as well as the arginine epsilon-amino group.
  • tyrosyl residues may be made, with particular interest in introducing spectral labels into tyrosyl residues by reaction with aromatic diazonium compounds or tetranitromethane. Most commonly, N-acetylimidizole and tetranitromethane are used to form O-acetyl tyrosyl species and 3-nitro derivatives, respectively.
  • Tyrosyl residues are iodinated using 125 I or 131 I to prepare labeled proteins for use in radioimmunoassay.
  • Carboxyl side groups are selectively modified by reaction with carbodiimides (R-N.dbd.C.dbd.N-R'), where R and R' are different alkyl groups, such as 1-cyclohexyl-3-(2-morpholinyl-4-ethyl) carbodiimide or 1-ethyl-3-(4-azonia-4,4-dimethylpentyl) carbodiimide.
  • aspartyl and glutamyl residues are converted to asparaginyl and glutaminyl residues by reaction with ammonium ions.
  • Glutaminyl and asparaginyl residues are frequently deamidated to the corresponding glutamyl and aspartyl residues, respectively. These residues are deamidated under neutral or basic conditions. The deamidated form of these residues falls within the scope of this invention.
  • Another type of covalent modification involves chemically or enzymatically coupling glycosides to the specific binding agent or antibody. These procedures are advantageous in that they do not require production of the specific binding agent or antibody in a host cell that has glycosylation capabilities for N- or O-linked glycosylation.
  • the sugar(s) may be attached to (a) arginine and histidine, (b) free carboxyl groups, (c) free sulfhydryl groups such as those of cysteine, (d) free hydroxyl groups such as those of serine, threonine, or hydroxyproline, (e) aromatic residues such as those of phenylalanine, tyrosine, or tryptophan, or (f) the amide group of glutamine.
  • Removal of any carbohydrate moieties present on the specific binding agent or antibody may be accomplished chemically or enzymatically.
  • Chemical deglycosylation requires exposure of the specific binding agent or antibody to the compound trifluoromethanesulfonic acid, or an equivalent compound. This treatment results in the cleavage of most or all sugars except the linking sugar (N-acetylglucosamine or N-acetylgalactosamine), while leaving the specific binding agent or antibody intact.
  • Chemical deglycosylation is described by Hakimuddin, et al. Arch. Biochem. Biophys. 259: 52 (1987 ) and by Edge et al. Anal. Biochem., 118: 131 (1981 ).
  • Enzymatic cleavage of carbohydrate moieties on a specific binding agent or antibody can be achieved by the use of a variety of endo- and exo-glycosidases as described by Thotakura et al. Meth. Enzymol. 138: 350 (1987 ).
  • Another type of covalent modification of the specific binding agent or antibody comprises linking the specific binding agent or antibody to one of a variety of nonproteinaceous polymers, e.g., polyethylene glycol, polypropylene glycol, polyoxyethylated polyols, polyoxyethylated sorbitol, polyoxyethylated glucose, polyoxyethylated glycerol, polyoxyalkylenes, or polysaccharide polymers such as dextran.
  • nonproteinaceous polymers e.g., polyethylene glycol, polypropylene glycol, polyoxyethylated polyols, polyoxyethylated sorbitol, polyoxyethylated glucose, polyoxyethylated glycerol, polyoxyalkylenes, or polysaccharide polymers such as dextran.
  • nonproteinaceous polymers e.g., polyethylene glycol, polypropylene glycol, polyoxyethylated polyols, poly
  • Treatment is an intervention performed with the intention of preventing the development or altering the pathology of a disorder. Accordingly, “treatment” refers to both therapeutic treatment and prophylactic or preventative measures. Those in need of treatment include those already with the disorder as well as those in which the disorder is to be prevented.
  • mammal for purposes of treatment refers to any animal classified as a mammal, including humans, domestic and farm animals, and zoo, sports, or pet animals, such as dogs, horses, cats, cows, etc. Preferably, the mammal is human.
  • the phrase "therapeutically effective amount” is meant to refer to an amount of therapeutic or prophylactic humanized c-Kit antibody that provides a reduction in mast cell or progenitor cell number and/or activity, reduction in fibroid elements or their precursors, or that provides a reduction in the severity or progression of symptoms associated with c-kit associated disease (i.e. that provides "therapeutic efficacy”).
  • Mast cells and progenitor hematopoietic pluripotent stem cells are the primary cell types expressing c-Kit and thus it is contemplated that cells derived from HSC such as mast cells and that are involved in diseases can be treated with the compositions and methods of the invention.
  • fibrotic-reducing activity is meant to refer to the ability to inhibit, fully or partially and reverse inflammation resulting from immune system activation and fibrosis.
  • fibrotic disease or disorder refers to conditions involving fibrosis in one or more tissues.
  • fibrosis refers to abberant formation or development of excess fibrous connective tissue in an organ or tissue as a reactive process, as opposed to formation of fibrous tissue as a normal constituent or healing of an organ or tissue. Fibrosis is characterized by fibroblast accumulation and collagen deposition in excess of normal deposition in any particular tissue.
  • fibrosis is used synonymously with "aberrant healing, involving mesenchymal-fibroblast cell transformation, excessive fibroblast proliferation, activity and deposition of collagens and other extracellular matrix proteins".
  • Fibroblasts are connective tissue cells, which are dispersed in connective tissue throughout the body. Fibroblasts secrete a nonrigid extracellular matrix containing type I and/or type III collagen. In response to an injury to a tissue, nearby fibroblasts or mesenchymal precursor cells in circulation migrate into the wound, may become alternatively activated under the influence of other cells such as mast cells and their mediators, proliferate, and produce large amounts of collagenous extracellular matrix.
  • Collagen is a fibrous protein rich in glycine and proline that is a major component of the extracellular matrix and connective tissue, cartilage, and bone. Collagen molecules are triple-stranded helical structures called ⁇ -chains, which are wound around each other in a ropelike helix. Collagen exists in several forms or types; of these, type I, the most common, is found in skin, tendon, and bone; and type III is found in skin, blood vessels, and internal organs.
  • Mast cell associated fibrotic diseases include pathological fibrosis or scarring (including endocardial sclerosis), idiopathic interstitial fibrosis, interstitial pulmonary fibrosis, perimuscular fibrosis, Symmers' fibrosis, pericentral fibrosis, hepatitis, dermatofibroma, billary cirrhosis, alcoholic cirrhosis, acute pulmonary fibrosis, idiopathic pulmonary fibrosis, acute respiratory distress syndrome, kidney fibrosis/glomerulonephritis, kidney fibrosis/diabetic nephropathy, scleroderma/systemic, scleroderma/local, keloids, hypertrophic scars, severe joint adhesions/arthritis, myelofibrosis, corneal scarring, cystic fibrosis, muscular dystrophy (duchenne's), cardiac fibrosis, muscular fibrosis/retinal separation, esoph
  • fibrotic disorders may be induced or initiated by surgery, including scar revision/plastic surgeries, glaucoma, cataract fibrosis, corneal scarring, joint adhesions, graft vs. host disease, tendon surgery, nerve entrapment, dupuytren's contracture, OB/GYN adhesions/fibrosis, pelvic adhesions, peridural fibrosis, restenosis. It is also contemplated that fibrotic conditions where deposition of fibronectin is a causative factor can be treated according to the invention.
  • Idiopathic pulmonary fibrosis, bleomycin lung, cystic fibrosis, and glomerular nephropathy, including disease characterized by Fn deposits in the kidneys ultimately leading to renal failure are examples of conditions which can also be treated in accordance with the present invention.
  • Inflammation involving activation of the immune system and where mast cells secrete inflammatory cytokines such as TNF, and can activate and directly interact with lymphocytes can also be treated in accordance with the present invention.
  • Scleroderma is believed to be an autoimmune disease of the connective tissue resulting in a fibrotic disorder characterized by a thickening and induration of the skin caused by the overproduction of new collagen by fibroblasts in skin and other organs. Scleroderma may occur as a local or systemic disease involving a number of organs. Scleroderma is also referred to as systemic sclerosis. The development of scleroderma pathologies is associated with increased mast cell numbers in the affected disease tissues/organs.
  • Systemic sclerosis is characterized by formation of hyalinized and thickened collagenous fibrous tissue, with thickening of the skin and adhesion to underlying tissues, especially of the hands and face.
  • the disease may also be characterized by dysphagia due to loss of peristalsis and submucosal fibrosis of the esophagus, dyspnea due to pulmonary fibrosis, myocardial fibrosis, and renal vascular changes. ( Stedman's Medical Dictionary, 26th Edition, Williams & Wilkins, 1995 )).
  • Pulmonary fibrosis affects 30 to 70% of scleroderma patients, often resulting in restrictive lung disease ( Atamas et al.
  • Some patients have an overlap of scleroderma and other connective tissue diseases, such as rheumatoid arthritis, systemic lupus erythematosus, and polymyositis.
  • scleroderma When features of scleroderma are present along with features of polymyositis and systemic lupus erythematosus, the condition is referred to as mixed connective tissue disease (MCTD).
  • MCTD mixed connective tissue disease
  • mast cell associated disorder suitable for treatment according to the invention is urticaria pigmentosa.
  • This disorder presents characteristic skin lesions that are single or multiple pigmented macules or nodules that urticate on rubbing and contain large numbers of mast cells.
  • associated dermatitis inflammation of skin
  • erythema, edema, papular eruptions and pruritus may be present in both human and animal dermatitides, all of which are treatable according to the invention.
  • Mastocytosis is in many cases a neoplastic disease and involves new or abnormal mast cell growth and may be a consequence of elevated SCF autocrine signaling or an activating c-Kit mutation.
  • Mastocytosis may be limited or systemic involving multiple organs such as the bone marrow.
  • Mast cells release certain mediators, or chemicals, of which one is histamine, into the body in response to certain events.
  • People with systemic mastocytosis develop an increase in the number of mast cells, or they develop abnormally shaped mast cells, which may not function properly.
  • the mast cells fail to die off when they are supposed to, further increasing the total mast cell burden. When the mast cells degranulate and release their contents it can cause many acute and potentially serious conditions or diseases.
  • Mast cell disorders also include proliferative disorders resulting in localized disease such as solitary cutaneous mastocytoma up to the more severe disease of mast cell leukemia.
  • proliferative disorders resulting in localized disease such as solitary cutaneous mastocytoma up to the more severe disease of mast cell leukemia.
  • Examples include cutaneous mastocytoma, aggressive mastocytosis, indolent mastocytosis, mastocytosis with associated hematologic disorder, urticaria pigmentosa, telangiectasia macularis eruptiva perstans (tmep), systemic mast cell disease, mast cell leukemia, myeloid leukemia, systemic mastocytosis (with or without cutaneous manifestations such as urticaria pigmentosa), mast cell activation syndrome/disorder, and more common pediatric mast cell disorders such as solitary mastocytoma and diffuse cutaneous mastocytosis.
  • Mast cell activation syndrome or disorder is characterized by a normal or nearly normal number of mast cells. However, the mast cells are easily triggered to release their contents, which results in many of the same symptoms. The danger of anaphylaxis and shock is present with this disorder, but unlike proliferative disorders of mast cells, this syndrome may not have the potential to progress to a more aggressive or malignant stage.
  • disorders associated with mast cell degranulation can include abdominal pain, hives and rashes, anaphylaxis, inflammation of the esophagus, blood pressure changes and shock, intestinal cramping and bloating, bone pain (mild to severe/debilitating), itching, with and without rashes, chest pain, liver, spleen and other organ involvement, cognitive difficulties/brain fog, malabsorption, degenerative disc disease, migraine headaches, diarrhea, muscle pain, dizziness/vertigo/lightheadedness, nausea, faintness, osteoporosis/ osteopenia, fatigue, peripheral neuropathy and paresthesias, flushing, rapid heart rate, gastroesophageal reflux, and vomiting.
  • mast cell specific mediators such as histamine and corticosteroids which among their activities cause mast cell apoptosis.
  • Additional mast cell related diseases include histamine-mediated allergic reactions that can be treated by inhibiting chemokine-induced mast cell and basophil degranulation and release of histamine.
  • mast cell associated disorders or diseases which may be effectively treated with the subject methods and compositions also include, but are not limited to, contact dermatitis, atopic dermatitis, allergic dermatitis, eczematous dermatitis and dermatitis caused by insect bites or stings.
  • mast cell related indications suitable for treatment by the methods and compositions of the invention include pulmonary inflammatory conditions in interstitial lung diseases for example sarcoidosis, neonatal respiratory distress syndrome (RDS), bronchopulmonary dysplasia (BPD), and conditions characterized by an elevation in serum PLA2 activity, such as adult RDS (ARDS).
  • pulmonary inflammatory conditions in interstitial lung diseases for example sarcoidosis, neonatal respiratory distress syndrome (RDS), bronchopulmonary dysplasia (BPD), and conditions characterized by an elevation in serum PLA2 activity, such as adult RDS (ARDS).
  • RDS neonatal respiratory distress syndrome
  • BPD bronchopulmonary dysplasia
  • ARDS adult RDS
  • Mast cells have also been published to show roles in arthritis. Mast cells are increased in the inflamed synovial tissues of RA and OA patients, and Gleevec has been shown to cause mast cell apoptosis in synovial explants and human case studies show efficacy in RA patients. Mast cells have roles in septic shock, pancreatitis, collagen vascular diseases, acute renal failure, peritonitis, and autoimmune uveitis.
  • mast cells participate in the pathophysiology of multiple sclerosis. It is thought that mast cells in the brain release vasoactive amines which may cause demyelination. Histamine released from mast cells may alter blood vessel integrity and cause partial breakdown of the blood-brain barrier again implicated in the etiology of multiple sclerosis. Thus, it is contemplated that the methods and compositions of the invention are suitable for treatment or amelioration of the morbidity associated with multiple sclerosis.
  • C-kit is also expressed on certain non-immune cells such as melanocytes and intestinal cells aswell as spermatocytes.
  • the invention may have utility in the treatment of melanoma and GIST and may have utility as a male contraceptive.
  • the anti-c-Kit specific binding agents or antibodies used in the practice of a method of the invention may be formulated into pharmaceutical compositions comprising a carrier suitable for the desired delivery method.
  • Suitable carriers include any material which, when combined with the anti-c-Kit specific binding and neutral antagonist agent or antibody, retains the high-affinity binding and potency at c-Kit and is nonreactive with the subject's immune systems. Examples include, but are not limited to, any of a number of standard pharmaceutical carriers such as sterile phosphate buffered saline solutions, bacteriostatic water, and the like.
  • aqueous carriers may be used, e.g., water, buffered water, 0.4% saline, 0.3% glycine and the like, and may include other proteins for enhanced stability, such as albumin, lipoprotein, globulin, etc., subjected to mild chemical modifications or the like.
  • Exemplary antibody concentrations in the formulation may range from about 0.1 mg/ml to about 180 mg/ml or from about 0.1 mg/mL to about 50 mg/mL, or from about 0.5 mg/mL to about 25 mg/mL, or alternatively from about 2 mg/mL to about 10 mg/mL.
  • An aqueous formulation of the antibody may be prepared in a pH-buffered solution, for example, at pH ranging from about 4.5 to about 6.5, or from about 4.8 to about 5.5, or alternatively about 5.0.
  • buffers that are suitable for a pH within this range include acetate (e.g. sodium acetate), succinate (such as sodium succinate), gluconate, histidine, citrate and other organic acid buffers.
  • the buffer concentration can be from about 1 mM to about 200 mM, or from about 10 mM to about 60 mM, depending, for example, on the buffer and the desired isotonicity of the formulation.
  • a tonicity agent which may also stabilize the antibody, may be included in the formulation.
  • exemplary tonicity agents include polyols, such as mannitol, sucrose or trehalose.
  • the aqueous formulation is isotonic, although hypertonic or hypotonic solutions may be suitable.
  • concentrations of the polyol in the formulation may range from about 1% to about 15% w/v.
  • a surfactant may also be added to the antibody formulation to reduce aggregation of the formulated antibody and/or minimize the formation of particulates in the formulation and/or reduce adsorption.
  • exemplary surfactants include nonionic surfactants such as polysorbates (e.g. polysorbate 20, or polysorbate 80) or poloxamers (e.g. poloxamer 188).
  • Exemplary concentrations of surfactant may range from about 0.001% to about 0.5%, or from about 0.005% to about 0.2%, or alternatively from about 0.004% to about 0.01% w/v.
  • the formulation contains the above-identified agents (i.e. antibody, buffer, polyol and surfactant) and is essentially free of one or more preservatives, such as benzyl alcohol, phenol, m-cresol, chlorobutanol and benzethonium Cl.
  • a preservative may be included in the formulation, e.g., at concentrations ranging from about 0.1% to about 2%, or alternatively from about 0.5% to about 1%.
  • One or more other pharmaceutically acceptable carriers, excipients or stabilizers such as those described in Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980 ) may be included in the formulation provided that they do not adversely affect the desired characteristics of the formulation.
  • Acceptable carriers, excipients or stabilizers are nontoxic to recipients at the dosages and concentrations employed and include; additional buffering agents; cosolvents; antoxidants including ascorbic acid and methionine; chelating agents such as EDTA; metal complexes (e.g. Zn-protein complexes); biodegradable polymers such as polyesters; and/or salt-forming counterions such as sodium.
  • Therapeutic formulations of the antibody are prepared for storage by mixing the antibody having the desired degree of purity with optional physiologically acceptable carriers, excipients or stabilizers ( Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980 )), in the form of lyophilized formulations or aqueous solutions.
  • Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine,
  • a suitable formulation of the claimed invention contains an isotonic buffer such as a phosphate, acetate, or TRIS buffer in combination with a tonicity agent such as a polyol, Sorbitol, sucrose or sodium chloride which tonicifies and stabilizes.
  • a tonicity agent such as a polyol, Sorbitol, sucrose or sodium chloride which tonicifies and stabilizes.
  • a tonicity agent is 5% Sorbitol or sucrose.
  • the formulation could optionally include a surfactant such as to prevent aggregation and for stabilization at 0.01 to 0.02% wt/vol.
  • the pH of the formulation may range from 4.5-6.5 or 4.5 to 5.5.
  • Other exemplary descriptions of pharmaceutical formulations for antibodies may be found in US 2003/0113316 and US patent no. 6,171,586 .
  • the formulation herein may also contain more than one active compound as necessary for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other.
  • active compound preferably those with complementary activities that do not adversely affect each other.
  • it may be desirable to further provide an immunosuppressive agent.
  • Such molecules are suitably present in combination in amounts that are effective for the purpose intended.
  • the active ingredients may also be entrapped in microcapsule prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsule and poly-(methylmethacylate) microcapsule, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions.
  • colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • Suspensions and crystal forms of antibodies are also contemplated. Methods to make suspensions and crystal forms are known to one of skill in the art.
  • compositions to be used for in vivo administration must be sterile.
  • the compositions of the invention may be sterilized by conventional, well known sterilization techniques. For example, sterilization is readily accomplished by filtration through sterile filtration membranes.
  • the resulting solutions may be packaged for use or filtered under aseptic conditions and lyophilized, the lyophilized preparation being combined with a sterile solution prior to administration.
  • a lyophilization cycle is usually composed of three steps: freezing, primary drying, and secondary drying; Williams and Polli, Journal of Parenteral Science and Technology, Volume 38, Number 2, pages 48-59 (1984 ).
  • freezing step the solution is cooled until it is adequately frozen.
  • Bulk water in the solution forms ice at this stage.
  • the ice sublimes in the primary drying stage, which is conducted by reducing chamber pressure below the vapor pressure of the ice, using a vacuum.
  • sorbed or bound water is removed at the secondary drying stage under reduced chamber pressure and an elevated shelf temperature.
  • the process produces a material known as a lyophilized cake. Thereafter the cake can be reconstituted prior to use.
  • Excipients have been noted in some cases to act as stabilizers for freeze-dried products; Carpenter et al., Developments in Biological Standardization, Volume 74, pages 225-239 (1991 ).
  • known excipients include polyols (including mannitol, sorbitol and glycerol); sugars (including glucose and sucrose); and amino acids (including alanine, glycine and glutamic acid).
  • polyols and sugars are also often used to protect polypeptides from freezing and drying-induced damage and to enhance the stability during storage in the dried state.
  • sugars in particular disaccharides, are effective in both the freeze-drying process and during storage.
  • Other classes of molecules including mono- and di-saccharides and polymers such as PVP, have also been reported as stabilizers of lyophilized products.
  • the pharmaceutical formulation and/or medicament may be a powder suitable for reconstitution with an appropriate solution as described above.
  • these include, but are not limited to, freeze dried, rotary dried or spray dried powders, amorphous powders, granules, precipitates, or particulates.
  • the formulations may optionally contain stabilizers, pH modifiers, surfactants, bioavailability modifiers and combinations of these.
  • Sustained-release preparations may be prepared. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the antibody, which matrices are in the form of shaped articles, e.g., films, or microcapsule. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or poly(vinylalcohol)), polylactides ( U.S. Patent No.
  • copolymers of L-glutamic acid and y ethyl-L-glutamate non-degradable ethylene-vinyl acetate
  • degradable lactic acid-glycolic acid copolymers such as the Lupron DepotTM (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate)
  • poly-D-(-)-3-hydroxybutyric acid While polymers such as ethylene-vinyl acetate and lactic acid-glycolic acid enable release of molecules for over 100 days, certain hydrogels release proteins for shorter time periods.
  • encapsulated antibodies When encapsulated antibodies remain in the body for a long time, they may denature or aggregate as a result of exposure to moisture at 37°C., resulting in a loss of biological activity and possible changes in immunogenicity. Rational strategies can be devised for stabilization depending on the mechanism involved. For example, if the aggregation mechanism is discovered to be intermolecular S--S bond formation through thio-disulfide interchange, stabilization may be achieved by modifying sulfhydryl residues, lyophilizing from acidic solutions, controlling moisture content, using appropriate additives, and developing specific polymer matrix compositions.
  • the formulations of the invention may be designed to be short-acting, fast-releasing, long-acting, or sustained-releasing as described herein.
  • the pharmaceutical formulations may also be formulated for controlled release or for slow release.
  • Specific dosages may be adjusted depending on conditions of disease, the age, body weight, general health conditions, sex, and diet of the subject, dose intervals, administration routes, excretion rate, and combinations of drugs. Any of the above dosage forms containing effective amounts are well within the bounds of routine experimentation and therefore, well within the scope of the instant invention.
  • the specific binding agent or antibody is administered by any suitable means, including parenteral, subcutaneous, intraperitoneal, intrapulmonary, and intranasal, and, if desired for local treatment, intralesional administration.
  • Parenteral infusions include intravenous, intraarterial, intraperitoneal, intramuscular, intradermal or subcutaneous administration.
  • the specific binding agent or antibody is suitably administered by pulse infusion, particularly with declining doses of the specific binding agent or antibody.
  • the dosing is given by injections, most preferably intravenous or subcutaneous injections, depending in part on whether the administration is brief or chronic.
  • Other administration methods are contemplated, including topical, particularly transdermal, transmucosal, rectal, oral or local administration e.g.
  • the specific binding agent or antibody of the invention is administered intravenously in a physiological solution at a dose ranging between 0.01 mg/kg to 100 mg/kg at a frequency ranging from daily to weekly to monthly (e.g. every day, every other day, every third day, or 2, 3, 4, 5, or 6 times per week), preferably a dose ranging from 0.1 to 45 mg/kg, 0.1 to 15 mg/kg or 0.1 to 10 mg/kg at a frequency of 2 or 3 times per week, or up to 45mg/kg once a month.
  • the antibodies of the invention also may be concurrently administered with other anti-inflammatory therapeutic agents.
  • Concurrent administration includes administration of the two different therapeutic agents at different times and at different routes, as long as there is some overlap in the time during which the agents are exerting their therapeutic effects.
  • Exemplary anti-c-Kit agents known in the art include Imatinib Mesylate (GleevecTM). It should be noted that Imatinib Mesylate also antagonizes signaling from the Abl tyrosine kinase and therefore is not a specific c-Kit inhibitor.
  • SR-1 was humanized by a straight CDR graft, with surprisingly no back mutations required to maintain affinity though it remained to be demonstrated desired functional activity.
  • the light chain acceptor sequence was the VK4 B3 germline sequence with JK2 as the closest J region.
  • Isotype switching was done to produce human IgG2, IgG1, IgG4P and aglycosylated IgG1 forms of the humanized antibody.
  • the N-linked glycosylation consensus site was removed from the human IgG1 constant region sequence by mutation of a single residue from asparagine to glutamic acid at position 297 (Kabat numbering).
  • the humanized SR-1 in the aglycosylated IgG1 (hSR-1 aIgG1) form binds in a desired manner with higher affinity to the membrane c-Kit compared to soluble c-kit, and is a highly potent neutral antagonist of SCF and mediating no agonism of c-Kit directly in all cell based assays tested using proximal and distal read-outs of c-Kit signaling.
  • the aglycosylated IgG1 isotype was chosen to avoid effector function and cell killing through bystander effects. This antibody showed an unexpected and desired half-life, nonlinear PK and saturable target-mediated antibody elimination in monkeys. It also depletes mast cells in vivo as expected.
  • SR-1 stem cell factor
  • SCF stem cell factor
  • the human megakaryoblastic cell line UT-7 is dependent on SCF for survival and the removal of SCF or its inhibition results in rapid loss of viability and decreased proliferation.
  • This assay is suited for IC50 potency determination of SCF antagonists.
  • hSR-1 aIgG1 exhibited a mean IC50 of 35 pM.
  • hSR-1 aIgG1 potently inhibited SCF-mediated c-Kit phosphorylation and internalization in MO7e cells indicating that the antibody can block SCF-mediated c-Kit signaling events.
  • SR-1 being capable of intrinsically mediating c-Kit internalization and phosphorylation
  • no evidence of agonism was detected in the MO7e c-Kit receptor proximal phosphorylation read-out for hSR-1 aIgG1.
  • hSR-1 IgG2 the IgG2 antibody was slightly less potent and did not fully inhibit SCF-mediated c-Kit receptor internalization.
  • hSR-1 aIgG1 shows neutralization at 1.0 ug/mL of the synergistic effect of SCF on GM-CSF derived colony formation using primary isolated human CD34+, CD117+ (c-Kit) bone marrow cells. Consistent with our novel finding that hSR-1 aIgG1 did not mediate c-Kit internalization or phosphorylation, and no intrinsic agonistic survival activity of hSR-1 aIgG1 was observed up to 10 ug/mL concentration of the antibody in this assay. In fact, the antibody was able to inhibit survival below baseline.
  • hSR-1 aIgG1 inhibited SCF-dependent mast cell survival, conferred no survival signal to mast cells, did not mediate c-Kit receptor phosphorylation ( Figure 3 ) and showed no ability to mediate homotypic mast cell aggregation.
  • the hSR-1 IgG2 was able to block SCF-mast cell survival but itself showed partial agonist activity conferring a survival signal, mediating c-Kit receptor phosphorylation and resulted in a reproducible effect on mast cell clustering. No unexpected abnormalities were observed for hSR-1 aIGgl when this antibody was dosed in vivo up to 30 mg/kg once weekly for 4 weeks or up to 150 mg/kg subcutaneously once weekly for 2 weeks in non-human primates.
  • Fc ⁇ receptor I CD64
  • Fc ⁇ receptor II CD32
  • Fc ⁇ receptor III CD16.
  • binding of SR-1 IgG1 and IgG4P isotypes was detected presumably to the high affinity Fc ⁇ RI. Therefore no ADCC activity is predicted for hSR-1 aIgG1, and experimental data to date show no complement dependent cytotoxic cell death.
  • Aglycosylated chimeric mouse/human IgG1 antibodies have been reported to retain some effector function ( Hybridoma. 1991 Apr;10(2):211-7 ) and so these desired activities of hSR-1 aIgG1 are not expected.
  • a preliminary PK study was conducted to compare hSR-1 IgG2 and hSR-1 aIgG1 PK in male cynomolgus monkeys after a single IV or SC administration at 3 mg/kg.
  • Time profiles indicate a non-linear PK for both.
  • the concentrations decreased more rapidly at lower concentrations.
  • the two antibodies showed similar exposures, as measured by C 0 /C max and AUC 0-tlast , after a single IV or SC administration in cynomolgus monkeys.
  • the serum clearance was approximately ⁇ 0.3 mL/hr/kg for both humanized antibodies.
  • the bioavailability was approximately 82% and 69% for the hSR-1 aIgG1 and hSR-1 IgG2 humanized SR-1 versions, respectively, after SC dosing.
  • the minimal effective dose in the wound PD model of mast cell expansion is ⁇ 0.3 mg/Kg administered once weekly for 2 weeks in monkeys. Based on a body surface area-based dose conversion, the minimal effective dose in human is projected to be ⁇ 0.1 mg/Kg with an equivalent dosing regiment.
  • this is a preliminary estimation as the PK and the pharmacodynamic relationship between the degree and duration of c-kit inhibition in human by hSR-1 aIgG1 and clinical endpoints are unknown at this time. A more accurate projection will be made when more pharmacokinetic and pharmacodynamic data are available.
  • mast cell MCt expressing tryptase and lacking chymase are localized primarily to mucosal tissues such as the lung and colon, and this subtype has been detected in the skin and at higher levels of some scleroderma patients suggesting possible alternative activation of mast cells in this condition.
  • the mast cell MCtc expressing both tryptase and chymase are also colocalized in some of these tissues and similarly have been associated with scleroderma and other fibrotic conditions.
  • both subtypes would represent the primary targets for a c-Kit inhibitor in diseases involving mucosal and connective tissues (eg. IPF, SSc, asthma, RA and IBD).
  • the therapeutic would also need to be highly potent, efficacious and have a good volume of distribution and PK since mast cells are generally long-lived and are tissue-resident. Moreover mast cells are largely quiescent until activated to degranulate and de novo synthesize mediators where they then play a key role in the inflammatory response.
  • the aims for the in vivo studies were to demonstrate depletion of basal mucosal and connective tissue mast cells such as present in the lung and colon and to determine effects on hematopoiesis and effects on precursor cells as well as an impact on erythropoiesis, melanogenesis and spermatogenesis (therefore utility in male contraception) following sustained and high fractional inhibition of c-Kit.
  • the SR-1 monoclonal antibody was selected based on its equivalent functional potency at human and monkey c-Kit in the CD34+ bone marrow cell CFU assay (inhibition at 1.0 ug/mL), and its monkey PK.
  • SR-1 was administered at doses ranging from 3 mg/Kg to 30 mg/Kg once weekly for 4 weeks.
  • basal colon mast cells were shown to be maximally depleted after 2 doses by day 14 (C trough >800-fold the cell IC50), and thus day 14 was chosen as the time point to determine the pharmacological activity of c-Kit antagonists on basal colon mast cells.
  • basal pulmonary mast cells were evaluated on day 28 at the time of necropsy and the termination of the study.
  • the multi-kinase inhibitor Gleevec which target primarily BCR-ABL, PDGFR and c-Kit has as its primary pharmacological effect myelosuppression, and severe grade 3-4 anemias and thrombocytopenias have been reported in GIST patients ( Hensley ML, et al., Semin. Hematol. 2003, Apr; 40(2 Suppl 2):21-5 ).
  • SR-1 was used in dose-ranging studies from 3 to 30 mg/Kg administered once weekly for 4 weeks to determine the exposures required to inhibit hematopoiesis, spermatogenesis and active melanogenesis.
  • a full blood panel including cell differentials was performed on blood samples taken at baseline, day 4, 7, 14, 21 and 28 after the start of the study. Analysis of freshly isolated blood samples was performed at the Queen Elizabeth Hospital Clinical Hematology lab, Barbados.
  • SR-1 showed dose-dependent inhibition of spermatogenesis from 0.3-30 mg/Kg.
  • the 0.3 mg/Kg once weekly dose is less than the maximal effect observed at the higher doses, and lower dose ranging studies are required to define the ED50.
  • the exposures achieved are 7-fold the UT-7 IC50 which is the exposure required for maximal efficacy in the wound model of mast cell expansion.
  • PK extrapolation suggested that the antibody may likely clear 1 month after the last dose, but 9 months was selected as a conservative time point to assess recovery.
  • Normal spermatogenesis was found in all dosed animals at 9 months demonstrating that use of a hSR-1 aIgG1-like molecule is useful as a male contraceptive.
  • Humanized anti-c-Kit aglycosylated IgG1 (hSR-1 aIgG1) antibody is a highly potent and specific antibody that is neutralizing in all cell based assays tested using proximal and distal read-outs of c-Kit signaling. Intrinsically it does not mediate c-Kit receptor internalization or phosphorylation as reported for the parent murine monoclonal SR-1 antibody.
  • the selection of the aglycosylated IgG1 isotype over humanized IgG1, IgG2 and IgG4 isotypes would not have been predicted based on standard approaches.
  • hSR-1 aIgG1 was chosen empirically via novel experimentation to show that it exhibited the appropriate pharmacological characteristics at the membrane c-Kit receptor, avoid agonist activity at c-Kit and on mast cells, lacking effector function and cell killing through bystander effects.
  • This antibody showed good s.c. bioavailability and half-life, nonlinear PK and saturable target-mediated antibody elimination and mast cell depletion in monkeys. These data would predict a suitable human efficacious mast cell depleting dose.
  • the minimal efficacious dose of the parent mouse SR-1 monoclonal in the monkey wound PD model is ⁇ 0.3 mg/Kg (C trough > 7-fold the cell IC50), and at slightly higher exposure (C trough > 800-fold the cell IC50) was efficacious as well in depleting basal skin, colon and lung mast cells.
  • exposure levels greater than >8000-fold the cell IC50 is required before qualitative impacts on hair pigmentation in newly regrown hair can be observed.
  • Inhibition of hair pigmentation in newly growing hair has been reported with multi-kinase inhibitors and a c-Kit antibody in rodent and the hSR-1 aIgG1 may have utility in diseases associated with excessive melanocyte activity. The effect is reversible upon cessation of treatment.
  • a sub-maximal effect on inhibition of spermatogenesis was shown at levels >7-fold the cell IC50, the exposure that conferred maximal efficacy in the wound PD model.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Virology (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Oncology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Pulmonology (AREA)
  • Hematology (AREA)
  • Communicable Diseases (AREA)
  • Rheumatology (AREA)
  • Biomedical Technology (AREA)
  • Diabetes (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Pain & Pain Management (AREA)
  • Dermatology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Mycology (AREA)
  • Hospice & Palliative Care (AREA)
  • Otolaryngology (AREA)
EP07756072.0A 2006-04-24 2007-04-24 Humanized c-kit antibody Active EP2010571B1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP18181319.7A EP3453724A1 (en) 2006-04-24 2007-04-24 Humanized c-kit antibody

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US79477106P 2006-04-24 2006-04-24
PCT/US2007/010155 WO2007127317A2 (en) 2006-04-24 2007-04-24 Humanized c-kit antibody

Related Child Applications (2)

Application Number Title Priority Date Filing Date
EP18181319.7A Division EP3453724A1 (en) 2006-04-24 2007-04-24 Humanized c-kit antibody
EP18181319.7A Division-Into EP3453724A1 (en) 2006-04-24 2007-04-24 Humanized c-kit antibody

Publications (2)

Publication Number Publication Date
EP2010571A2 EP2010571A2 (en) 2009-01-07
EP2010571B1 true EP2010571B1 (en) 2018-10-17

Family

ID=38543531

Family Applications (2)

Application Number Title Priority Date Filing Date
EP18181319.7A Pending EP3453724A1 (en) 2006-04-24 2007-04-24 Humanized c-kit antibody
EP07756072.0A Active EP2010571B1 (en) 2006-04-24 2007-04-24 Humanized c-kit antibody

Family Applications Before (1)

Application Number Title Priority Date Filing Date
EP18181319.7A Pending EP3453724A1 (en) 2006-04-24 2007-04-24 Humanized c-kit antibody

Country Status (22)

Country Link
US (3) US7915391B2 (no)
EP (2) EP3453724A1 (no)
JP (3) JP5094842B2 (no)
KR (1) KR101110547B1 (no)
CN (1) CN101472949A (no)
AR (1) AR060583A1 (no)
AU (1) AU2007243318B2 (no)
BR (1) BRPI0712920A2 (no)
CA (1) CA2648882C (no)
CR (1) CR10444A (no)
EA (1) EA015923B1 (no)
ES (1) ES2705480T3 (no)
IL (1) IL194776A (no)
MX (1) MX2008013640A (no)
MY (1) MY153710A (no)
NO (1) NO20084878L (no)
NZ (2) NZ594968A (no)
PE (1) PE20080694A1 (no)
TW (1) TWI395754B (no)
UA (1) UA95478C2 (no)
WO (1) WO2007127317A2 (no)
ZA (1) ZA200809065B (no)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11041022B2 (en) 2018-11-26 2021-06-22 Forty Seven, Inc. Humanized antibodies against c-Kit

Families Citing this family (189)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DK2006381T3 (en) 2006-03-31 2016-02-22 Chugai Pharmaceutical Co Ltd PROCEDURE FOR REGULATING ANTIBODIES BLOOD PHARMACOKINETICS
TWI395754B (zh) * 2006-04-24 2013-05-11 Amgen Inc 人類化之c-kit抗體
EP3127921A1 (en) 2007-09-26 2017-02-08 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substition in cdr
RU2571225C2 (ru) 2008-04-11 2015-12-20 Чугаи Сейяку Кабусики Кайся Антигенсвязывающая молекула, способная к многократному связыванию двух или более молекул антигена
WO2010028288A2 (en) 2008-09-05 2010-03-11 Aueon, Inc. Methods for stratifying and annotating cancer drug treatment options
CA2778105C (en) 2009-10-23 2019-04-02 Amgen Inc. Vial adapter and system
EP2619329B1 (en) 2010-09-24 2019-05-22 The Board of Trustees of The Leland Stanford Junior University Direct capture, amplification and sequencing of target dna using immobilized primers
CA2817216A1 (en) 2010-11-08 2012-05-18 F. Hoffmann-La Roche Ag Subcutaneously administered anti-il-6 receptor antibody
BR112013013354A2 (pt) 2010-11-30 2020-10-06 Chugai Seiyaku Kabushiki Kaisha molécula de ligação ao antígeno capaz de se ligar a uma pluralidade de moléculas de antígeno repetidamente
KR102119187B1 (ko) 2011-01-10 2020-06-08 더 리젠츠 오브 더 유니버시티 오브 미시간 줄기 세포 인자 억제제
EP3763740A1 (en) 2011-01-26 2021-01-13 Celldex Therapeutics, Inc. Anti-kit antibodies and uses thereof
KR102147548B1 (ko) 2011-02-25 2020-08-24 추가이 세이야쿠 가부시키가이샤 FcγRIIb 특이적 Fc 항체
MX341790B (es) 2011-03-31 2016-09-02 Amgen Inc Adaptador de viales y sistema.
CA3021845C (en) 2011-04-20 2022-03-29 Amgen Inc. Autoinjector apparatus
AR086044A1 (es) 2011-05-12 2013-11-13 Imclone Llc Anticuerpos que se unen especificamente a un dominio extracelular de c-kit y usos de los mismos
WO2013033008A2 (en) 2011-08-26 2013-03-07 Merrimack Pharmaceuticals, Inc. Tandem fc bispecific antibodies
TW201817745A (zh) 2011-09-30 2018-05-16 日商中外製藥股份有限公司 具有促進抗原清除之FcRn結合域的治療性抗原結合分子
WO2013047748A1 (ja) 2011-09-30 2013-04-04 中外製薬株式会社 複数の生理活性を有する抗原の消失を促進する抗原結合分子
TR201908439T4 (tr) 2011-10-14 2019-07-22 Amgen Inc Enjektör ve montaj yöntemi.
EA034989B1 (ru) 2011-10-28 2020-04-15 Тева Фармасьютикал Австралия Пти Лтд Полипептидная конструкция для применения при лечении рака, включающая аттенуированный альфа-интерферон
CA3078979C (en) * 2011-10-31 2022-10-11 Genentech, Inc. Formulation comprising an anti-il13 antibody having extended stability
CN104080909A (zh) 2011-11-30 2014-10-01 中外制药株式会社 包含进入细胞内以形成免疫复合体的搬运体(载体)的药物
HUE049860T2 (hu) 2012-02-13 2020-11-30 Agency Science Tech & Res IL-ß-t neutralizáló, humán monoklonális antitestek
WO2013148232A1 (en) * 2012-03-27 2013-10-03 Genentech, Inc. Methods of prognosing, diagnosing and treating idiopathic pulmonary fibrosis
KR101384360B1 (ko) * 2012-05-04 2014-04-14 아주대학교산학협력단 Scf 또는 이의 수용체를 억제하는 물질을 포함하는 혈관 투과성 관련 질환의 치료 또는 예방용 조성물
BR112015001459B1 (pt) 2012-07-25 2023-02-14 Celldex Therapeutics, Inc Anticorpo isolado ou fragmento do mesmo, conjugado, usos dos mesmos, composição farmacêutica, polinucleotídeo, vetor, célula hospedeira, célula isolada, kit, método in vitro para inibir atividade da kit, método para produzir um anticorpo
NZ705370A (en) 2012-08-24 2018-06-29 Chugai Pharmaceutical Co Ltd Fcγriib-specific fc region variant
EP3072548B1 (en) 2012-11-21 2019-03-06 Amgen, Inc Drug delivery device
UY35148A (es) 2012-11-21 2014-05-30 Amgen Inc Immunoglobulinas heterodiméricas
JP2016510755A (ja) 2013-03-06 2016-04-11 メリマック ファーマシューティカルズ インコーポレーティッド 抗C−METタンデムFc二重特異性抗体
EP3593839A1 (en) 2013-03-15 2020-01-15 Amgen Inc. Drug cassette
US9708375B2 (en) 2013-03-15 2017-07-18 Amgen Inc. Inhibitory polypeptides specific to WNT inhibitors
BR112015021134A2 (pt) 2013-03-15 2017-10-10 Novartis Ag conjugados de fármaco e anticorpo
US10850037B2 (en) 2013-03-22 2020-12-01 Amgen Inc. Injector and method of assembly
CA2908350C (en) 2013-04-02 2023-08-08 Futa Mimoto Fc region variant
WO2015006554A1 (en) 2013-07-10 2015-01-15 The Regents Of The University Of Michigan Therapeutic antibodies and uses thereof
WO2015035215A1 (en) 2013-09-05 2015-03-12 Amgen Inc. Fc-containing molecules exhibiting predictable, consistent, and reproducible glycoform profiles
MX2016005315A (es) 2013-10-24 2016-08-11 Amgen Inc Sistema de administracion de farmacos con control sensible a la temperatura.
MX2016005312A (es) 2013-10-24 2016-08-11 Amgen Inc Inyector y metodo de montaje.
US9932410B2 (en) 2013-11-05 2018-04-03 Inserm (Institut National De La Sante Et De La Recherche Medicale Human neutralizing anti-kit antibodies and uses thereof
US10994112B2 (en) 2014-02-05 2021-05-04 Amgen Inc. Drug delivery system with electromagnetic field generator
ES2939760T3 (es) 2014-03-15 2023-04-26 Novartis Ag Tratamiento de cáncer utilizando un receptor quimérico para antígenos
CA3193070A1 (en) 2014-05-07 2015-11-12 Amgen Inc. Autoinjector with shock reducing elements
US10329556B2 (en) * 2014-05-13 2019-06-25 Bioatla, Llc Conditionally active biological proteins
MX2016014761A (es) 2014-05-16 2017-05-25 Amgen Inc Ensayo para detectar poblaciones celulares linfocitos t colaboradores 1 (th1) y linfocitos t colaboradores (th2).
EP3145543A4 (en) * 2014-05-23 2017-12-13 Celldex Therapeutics, Inc. Treatment of eosinophil or mast cell related disorders
US20170124285A1 (en) 2014-06-03 2017-05-04 Amgen Inc. Devices and methods for assisting a user of a drug delivery device
JP6532136B2 (ja) * 2014-06-09 2019-06-19 株式会社カネカ 抗体に結合する低分子化合物のスクリーニング方法
WO2016014530A1 (en) 2014-07-21 2016-01-28 Novartis Ag Combinations of low, immune enhancing. doses of mtor inhibitors and cars
MX2021014323A (es) 2014-10-14 2023-02-02 Amgen Inc Dispositivo de inyección de fármaco con indicadores visuales y audibles.
US20180334490A1 (en) 2014-12-03 2018-11-22 Qilong H. Wu Methods for b cell preconditioning in car therapy
EP3689394A1 (en) 2014-12-19 2020-08-05 Amgen Inc. Drug delivery device with live button or user interface field
WO2016100781A1 (en) 2014-12-19 2016-06-23 Amgen Inc. Drug delivery device with proximity sensor
WO2016126608A1 (en) 2015-02-02 2016-08-11 Novartis Ag Car-expressing cells against multiple tumor antigens and uses thereof
MX2017010466A (es) 2015-02-17 2018-06-06 Amgen Inc Dispositivo de administracion de farmacos con sujecion asistida de vacio y/o respuestas.
WO2016138434A1 (en) 2015-02-27 2016-09-01 Amgen Inc. Drug delivery device having a needle guard mechanism with a tunable threshold of resistance to needle guard movement
WO2016154588A1 (en) 2015-03-25 2016-09-29 Children's Hospital Medical Center Use of kit inhibitors to condition subjects for a hematopoietic stem cell (hsc) transplantation
WO2016172583A1 (en) 2015-04-23 2016-10-27 Novartis Ag Treatment of cancer using chimeric antigen receptor and protein kinase a blocker
EP3331913A1 (en) 2015-08-07 2018-06-13 Novartis AG Treatment of cancer using chimeric cd3 receptor proteins
WO2017039786A1 (en) 2015-09-02 2017-03-09 Amgen Inc. Syringe assembly adapter for a syringe
US10604577B2 (en) * 2015-10-22 2020-03-31 Allakos Inc. Methods and compositions for treating systemic mastocytosis
CN116334143A (zh) 2015-11-23 2023-06-27 诺华股份有限公司 优化的慢病毒转移载体及其用途
US11351308B2 (en) 2015-12-09 2022-06-07 Amgen Inc. Auto-injector with signaling cap
BR112018013074A2 (pt) 2015-12-30 2018-12-11 Novartis Ag terapias de célula efetora imune com eficácia real-çada
US11154661B2 (en) 2016-01-06 2021-10-26 Amgen Inc. Auto-injector with signaling electronics
US20200281973A1 (en) 2016-03-04 2020-09-10 Novartis Ag Cells expressing multiple chimeric antigen receptor (car) molecules and uses therefore
ES2814287T3 (es) 2016-03-15 2021-03-26 Amgen Inc Reducir la probabilidad de rotura de cristal en dispositivos de administración de fármaco
US11549099B2 (en) 2016-03-23 2023-01-10 Novartis Ag Cell secreted minibodies and uses thereof
CA3021027A1 (en) 2016-04-15 2017-10-19 Novartis Ag Compositions and methods for selective expression of chimeric antigen receptors
WO2017189089A1 (en) 2016-04-29 2017-11-02 Amgen Inc. Drug delivery device with messaging label
US11389588B2 (en) 2016-05-02 2022-07-19 Amgen Inc. Syringe adapter and guide for filling an on-body injector
WO2017197222A1 (en) 2016-05-13 2017-11-16 Amgen Inc. Vial sleeve assembly
US11238150B2 (en) 2016-05-16 2022-02-01 Amgen Inc. Data encryption in medical devices with limited computational capability
WO2017209899A1 (en) 2016-06-03 2017-12-07 Amgen Inc. Impact testing apparatuses and methods for drug delivery devices
KR20190038537A (ko) 2016-06-17 2019-04-08 마젠타 테라퓨틱스 인코포레이티드 Cd117+ 세포의 고갈을 위한 조성물 및 방법
EP3478342A1 (en) 2016-07-01 2019-05-08 Amgen Inc. Drug delivery device having minimized risk of component fracture upon impact events
SG11201900885VA (en) 2016-08-01 2019-02-27 Novartis Ag Treatment of cancer using a chimeric antigen receptor in combination with an inhibitor of a pro-m2 macrophage molecule
US20190328965A1 (en) 2016-08-17 2019-10-31 Amgen Inc. Drug delivery device with placement detection
EP3532127A1 (en) 2016-10-25 2019-09-04 Amgen Inc. On-body injector
WO2018111340A1 (en) 2016-12-16 2018-06-21 Novartis Ag Methods for determining potency and proliferative function of chimeric antigen receptor (car)-t cells
JOP20190155A1 (ar) 2016-12-21 2019-06-23 Novartis Ag مترافقات عقار جسم مضاد لإزالة خلايا جذعية مكونة للدم
JP2020503976A (ja) 2017-01-17 2020-02-06 アムジエン・インコーポレーテツド 注入デバイスならびに関連する使用および組立方法
JP7256744B2 (ja) 2017-01-20 2023-04-12 マジェンタ セラピューティクス インコーポレイテッド Cd137+細胞の枯渇のための組成物および方法
EP3574005B1 (en) 2017-01-26 2021-12-15 Novartis AG Cd28 compositions and methods for chimeric antigen receptor therapy
CN110312527A (zh) 2017-01-30 2019-10-08 小利兰·斯坦福大学托管委员会 用于干细胞移植的非基因毒性预处理方案
EP3577134A1 (en) 2017-01-31 2019-12-11 Novartis AG Treatment of cancer using chimeric t cell receptor proteins having multiple specificities
AU2018220538B2 (en) 2017-02-17 2023-12-14 Amgen Inc. Drug delivery device with sterile fluid flowpath and related method of assembly
EP3582829A1 (en) 2017-02-17 2019-12-25 Amgen Inc. Insertion mechanism for drug delivery device
US20200048359A1 (en) 2017-02-28 2020-02-13 Novartis Ag Shp inhibitor compositions and uses for chimeric antigen receptor therapy
AU2018231107B2 (en) 2017-03-06 2023-04-20 Amgen Inc. Drug delivery device with activation prevention feature
SG11201908058UA (en) 2017-03-07 2019-09-27 Amgen Inc Needle insertion by overpressure
CA3052310A1 (en) 2017-03-09 2018-09-13 Amgen Inc. Insertion mechanism for drug delivery device
JP2020513813A (ja) 2017-03-14 2020-05-21 アムジエン・インコーポレーテツド 細胞培養において産生される抗体の総非フコシル化グリコフォームの調節
FI3600491T3 (fi) 2017-03-28 2023-10-20 Amgen Inc Männänvarren ja ruiskukokoonpanon järjestelmä ja menetelmä
BR112019022912A2 (pt) 2017-05-05 2020-05-26 Allakos Inc. Métodos e composições para tratar doenças oculares alérgicas
WO2018226515A1 (en) 2017-06-08 2018-12-13 Amgen Inc. Syringe assembly for a drug delivery device and method of assembly
CN110709121B (zh) 2017-06-08 2022-06-24 安进公司 扭矩驱动式药物递送装置
WO2018229715A1 (en) 2017-06-16 2018-12-20 Novartis Ag Compositions comprising anti-cd32b antibodies and methods of use thereof
EP3641857A1 (en) 2017-06-22 2020-04-29 Amgen Inc. Device activation impact/shock reduction
MA49461A (fr) 2017-06-23 2020-04-29 Amgen Inc Dispositif électronique d'administration de médicament comprenant un bouchon activé par un ensemble commutateur
JP7408398B2 (ja) 2017-07-14 2024-01-05 アムジエン・インコーポレーテツド 二重ねじりばねシステムを有する針挿入後退システム
EP4292576A3 (en) 2017-07-21 2024-01-17 Amgen Inc. Gas permeable sealing member for drug container and methods of assembly
MA49677A (fr) 2017-07-25 2021-04-21 Amgen Inc Dispositif d'administration de médicament avec module d'engrenage et procédé d'assemblage associé
US11484648B2 (en) 2017-07-25 2022-11-01 Amgen Inc. Drug delivery device with container access system and related method of assembly
US20200164155A1 (en) 2017-08-09 2020-05-28 Amgen Inc. Hydraulic-pneumatic pressurized chamber drug delivery system
WO2019036181A1 (en) 2017-08-18 2019-02-21 Amgen Inc. BODY INJECTOR WITH STERILE ADHESIVE PATCH
US11103636B2 (en) 2017-08-22 2021-08-31 Amgen Inc. Needle insertion mechanism for drug delivery device
ES2939292T3 (es) 2017-10-04 2023-04-20 Amgen Inc Adaptador de flujo para dispositivo de administración de fármacos
ES2971450T3 (es) 2017-10-06 2024-06-05 Amgen Inc Dispositivo de administración de fármacos con conjunto de enclavamiento y procedimiento de montaje correspondiente
WO2019074579A1 (en) 2017-10-09 2019-04-18 Amgen Inc. DRUG DELIVERY DEVICE COMPRISING A DRIVE ASSEMBLY AND ASSEMBLY METHOD THEREOF
SG11202003415XA (en) 2017-10-18 2020-05-28 Novartis Ag Compositions and methods for selective protein degradation
CA3079897A1 (en) * 2017-10-24 2019-05-02 Magenta Therapeutics, Inc. Compositions and methods for the depletion of cd117+ cells
TW201922782A (zh) 2017-10-24 2019-06-16 美商麥珍塔治療學股份有限公司 去除cd117+細胞之組合物及方法
SG11202003177RA (en) 2017-10-25 2020-05-28 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
US20210040205A1 (en) 2017-10-25 2021-02-11 Novartis Ag Antibodies targeting cd32b and methods of use thereof
WO2019089798A1 (en) 2017-10-31 2019-05-09 Novartis Ag Anti-car compositions and methods
WO2019090079A1 (en) 2017-11-03 2019-05-09 Amgen Inc. System and approaches for sterilizing a drug delivery device
EP3707075A1 (en) 2017-11-06 2020-09-16 Amgen Inc. Fill-finish assemblies and related methods
US20200261648A1 (en) 2017-11-06 2020-08-20 Amgen Inc. Drug delivery device with placement and flow sensing
CA3079665A1 (en) 2017-11-10 2019-05-16 Amgen Inc. Plungers for drug delivery devices
SG11202003004RA (en) 2017-11-16 2020-04-29 Amgen Inc Door latch mechanism for drug delivery device
EA202092286A1 (ru) 2018-03-26 2021-03-18 Эмджен Инк. Общие афукозилированные гликоформы антител, полученные в культуре клеток
EP3784351A1 (en) 2018-04-27 2021-03-03 Novartis AG Car t cell therapies with enhanced efficacy
US20210396739A1 (en) 2018-05-01 2021-12-23 Novartis Ag Biomarkers for evaluating car-t cells to predict clinical outcome
WO2019227003A1 (en) 2018-05-25 2019-11-28 Novartis Ag Combination therapy with chimeric antigen receptor (car) therapies
US10835685B2 (en) 2018-05-30 2020-11-17 Amgen Inc. Thermal spring release mechanism for a drug delivery device
US11083840B2 (en) 2018-06-01 2021-08-10 Amgen Inc. Modular fluid path assemblies for drug delivery devices
US20210123075A1 (en) 2018-06-08 2021-04-29 Novartis Ag Compositions and methods for immunooncology
CN108822212B (zh) * 2018-06-08 2021-06-29 北京大学 选择性识别致癌性突变体kit itd的单克隆抗体及其应用
AR116109A1 (es) 2018-07-10 2021-03-31 Novartis Ag Derivados de 3-(5-amino-1-oxoisoindolin-2-il)piperidina-2,6-diona y usos de los mismos
WO2020023220A1 (en) 2018-07-24 2020-01-30 Amgen Inc. Hybrid drug delivery devices with tacky skin attachment portion and related method of preparation
US12042645B2 (en) 2018-07-24 2024-07-23 Amgen Inc. Delivery devices for administering drugs
MA53375A (fr) 2018-07-24 2021-06-02 Amgen Inc Dispositifs d'administration pour l'administration de médicaments
WO2020023336A1 (en) 2018-07-24 2020-01-30 Amgen Inc. Hybrid drug delivery devices with grip portion
MA53320A (fr) 2018-07-31 2021-11-03 Amgen Inc Ensemble de trajet de fluide pour dispositif d'administration de médicament
AU2019347710A1 (en) 2018-09-24 2021-02-04 Amgen Inc. Interventional dosing systems and methods
IL281469B2 (en) 2018-09-28 2024-08-01 Amgen Inc Assembling a memory alloy ejector activation assembly for a drug delivery device
CN112805048B (zh) 2018-10-02 2023-09-22 安进公司 具有内部力传递的用于药物递送的注射系统
MX2021003491A (es) 2018-10-05 2021-06-18 Amgen Inc Dispositivo de administracion de farmacos con indicador de dosis.
CA3112355A1 (en) 2018-10-15 2020-04-23 Amgen Inc. Drug delivery device having damping mechanism
WO2020081480A1 (en) 2018-10-15 2020-04-23 Amgen Inc. Platform assembly process for drug delivery device
US20200376135A1 (en) * 2018-10-23 2020-12-03 Magenta Therapeutics, Inc. Fc silenced antibody drug conjugates (adcs) and uses thereof
WO2020091981A1 (en) 2018-11-01 2020-05-07 Amgen Inc. Drug delivery devices with partial drug delivery member retraction
TWI831847B (zh) 2018-11-01 2024-02-11 美商安進公司 部分針頭縮回之藥物遞送裝置及其操作方法
MA54048A (fr) 2018-11-01 2022-02-09 Amgen Inc Dispositifs d'administration de médicament avec rétraction partielle de l'organe d'administration de médicament
EP3886869A4 (en) 2018-11-28 2022-07-06 Forty Seven, Inc. GENETICALLY MODIFIED CSPH RESISTANT TO ABLATIVE TREATMENT
MX2021007392A (es) 2018-12-20 2021-08-24 Novartis Ag Regimen de dosificacion y combinacion farmaceutica que comprende derivados de 3-(1-oxoisoindolin-2-il)piperidina-2,6-diona.
KR20210129671A (ko) 2019-02-15 2021-10-28 노파르티스 아게 3-(1-옥소-5-(피페리딘-4-일)이소인돌린-2-일)피페리딘-2,6-디온 유도체 및 이의 용도
BR112021015672A2 (pt) 2019-02-15 2021-10-05 Novartis Ag Derivados de 3-(1-oxoisoindolin-2-il)piperidina-2,6-diona substituída e usos dos mesmos
WO2020219742A1 (en) 2019-04-24 2020-10-29 Novartis Ag Compositions and methods for selective protein degradation
WO2020219482A1 (en) 2019-04-24 2020-10-29 Amgen Inc. Syringe sterilization verification assemblies and methods
EP4017560A2 (en) 2019-08-23 2022-06-29 Amgen, Inc Drug delivery device with configurable needle shield engagement components and related methods
US20220349898A1 (en) 2019-09-26 2022-11-03 Amgen Inc. Methods of producing antibody compositions
KR20210063782A (ko) * 2019-11-25 2021-06-02 주식회사 노벨티노빌리티 c-kit에 대한 항체 및 이의 용도
IL293834A (en) 2019-12-20 2022-08-01 Novartis Ag A combination of anti-tim-3 antibody mbg453 and anti, nis793 tgf-beta antibody with or without decitabine or anti-pd-1 antibody spratlizumab, for the treatment of myelofibrosis and myelodysplastic syndrome
CA3165735A1 (en) 2019-12-24 2021-07-01 Carna Biosciences, Inc. Diacylglycerol kinase modulating compounds
KR20220142484A (ko) 2020-02-14 2022-10-21 조운스 테라퓨틱스, 인크. Ccr8에 결합하는 항체 및 융합 단백질, 및 이의 용도
WO2021247892A1 (en) 2020-06-04 2021-12-09 Amgen Inc. Assessment of cleaning procedures of a biotherapeutic manufacturing process
CN116096862A (zh) 2020-06-11 2023-05-09 诺华股份有限公司 Zbtb32抑制剂及其用途
MX2022015852A (es) 2020-06-23 2023-01-24 Novartis Ag Regimen de dosificacion que comprende derivados de 3-(1-oxoisoindolin-2-il)piperidina-2,6-diona.
EP4188549A1 (en) 2020-08-03 2023-06-07 Novartis AG Heteroaryl substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
KR20230087539A (ko) 2020-10-15 2023-06-16 암젠 인크 항체 생산 방법에서의 상대적인 비결합 글리칸
WO2022081828A1 (en) * 2020-10-16 2022-04-21 The Board Of Trustees Of The Leland Stanford Junior University Hematopoietic stem cell engraftment with a combination of agents
TW202304979A (zh) 2021-04-07 2023-02-01 瑞士商諾華公司 抗TGFβ抗體及其他治療劑用於治療增殖性疾病之用途
AU2022267891A1 (en) 2021-04-27 2023-11-09 Novartis Ag Viral vector production system
WO2022246055A1 (en) 2021-05-21 2022-11-24 Amgen Inc. Method of optimizing a filling recipe for a drug container
WO2022261021A1 (en) 2021-06-07 2022-12-15 Amgen Inc. Using fucosidase to control afucosylation level of glycosylated proteins
JP2024520593A (ja) 2021-06-23 2024-05-24 ギリアード サイエンシーズ, インコーポレイテッド ジアシルグリエルコール(diacylglyercol)キナーゼ調節化合物
US11976072B2 (en) 2021-06-23 2024-05-07 Gilead Sciences, Inc. Diacylglycerol kinase modulating compounds
WO2022271650A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
EP4359411A1 (en) 2021-06-23 2024-05-01 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
CA3233279A1 (en) 2021-10-05 2023-04-13 Amgen Inc. Fc-gamma receptor ii binding and glycan content
KR20240091056A (ko) 2021-10-28 2024-06-21 길리애드 사이언시즈, 인코포레이티드 피리디진-3(2h)-온 유도체
CN118201941A (zh) 2021-10-29 2024-06-14 吉利德科学公司 Cd73化合物
CA3240527A1 (en) 2021-12-16 2023-06-22 Universitat Basel Discernible cell surface protein variants of cd117 for use in cell therapy
WO2023122615A1 (en) 2021-12-22 2023-06-29 Gilead Sciences, Inc. Ikaros zinc finger family degraders and uses thereof
WO2023122581A2 (en) 2021-12-22 2023-06-29 Gilead Sciences, Inc. Ikaros zinc finger family degraders and uses thereof
TW202340168A (zh) 2022-01-28 2023-10-16 美商基利科學股份有限公司 Parp7抑制劑
WO2023164305A1 (en) * 2022-02-28 2023-08-31 Jasper Therapeutics, Inc. Compositions and methods for depletion of diseased hematopoietic stem cells
EP4245756A1 (en) 2022-03-17 2023-09-20 Gilead Sciences, Inc. Ikaros zinc finger family degraders and uses thereof
TW202400138A (zh) 2022-04-21 2024-01-01 美商基利科學股份有限公司 Kras g12d調節化合物
WO2023215725A1 (en) 2022-05-02 2023-11-09 Fred Hutchinson Cancer Center Compositions and methods for cellular immunotherapy
WO2023214325A1 (en) 2022-05-05 2023-11-09 Novartis Ag Pyrazolopyrimidine derivatives and uses thereof as tet2 inhibitors
US20240116928A1 (en) 2022-07-01 2024-04-11 Gilead Sciences, Inc. Cd73 compounds
WO2024008910A1 (en) 2022-07-07 2024-01-11 Cimeio Therapeutics Ag Antibodies targeting cd117
WO2024040194A1 (en) 2022-08-17 2024-02-22 Capstan Therapeutics, Inc. Conditioning for in vivo immune cell engineering
WO2024089639A1 (en) 2022-10-26 2024-05-02 Novartis Ag Lentiviral formulations
US20240254118A1 (en) 2022-12-22 2024-08-01 Gilead Sciences, Inc. Prmt5 inhibitors and uses thereof
WO2024163905A1 (en) 2023-02-03 2024-08-08 Genzyme Corporation Hsc-specific antibody conjugated lipid nanoparticles and uses thereof

Family Cites Families (43)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US30985A (en) 1860-12-18 Thomas l
US3773919A (en) 1969-10-23 1973-11-20 Du Pont Polylactide-drug mixtures
US4179337A (en) 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
JPS6023084B2 (ja) 1979-07-11 1985-06-05 味の素株式会社 代用血液
US4640835A (en) 1981-10-30 1987-02-03 Nippon Chemiphar Company, Ltd. Plasminogen activator derivatives
JPS5896026A (ja) 1981-10-30 1983-06-07 Nippon Chemiphar Co Ltd 新規ウロキナ−ゼ誘導体およびその製造法ならびにそれを含有する血栓溶解剤
US4609546A (en) 1982-06-24 1986-09-02 Japan Chemical Research Co., Ltd. Long-acting composition
US4560655A (en) 1982-12-16 1985-12-24 Immunex Corporation Serum-free cell culture medium and process for making same
US4657866A (en) 1982-12-21 1987-04-14 Sudhir Kumar Serum-free, synthetic, completely chemically defined tissue culture media
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4767704A (en) 1983-10-07 1988-08-30 Columbia University In The City Of New York Protein-free culture medium
US4496689A (en) 1983-12-27 1985-01-29 Miles Laboratories, Inc. Covalently attached complex of alpha-1-proteinase inhibitor with a water soluble polymer
EP0206448B1 (en) 1985-06-19 1990-11-14 Ajinomoto Co., Inc. Hemoglobin combined with a poly(alkylene oxide)
US4766106A (en) 1985-06-26 1988-08-23 Cetus Corporation Solubilization of proteins for pharmaceutical compositions using polymer conjugation
GB8516415D0 (en) 1985-06-28 1985-07-31 Celltech Ltd Culture of animal cells
WO1987005330A1 (en) 1986-03-07 1987-09-11 Michel Louis Eugene Bergh Method for enhancing glycoprotein stability
US4927762A (en) 1986-04-01 1990-05-22 Cell Enterprises, Inc. Cell culture medium with antioxidant
US4791192A (en) 1986-06-26 1988-12-13 Takeda Chemical Industries, Ltd. Chemically modified protein with polyethyleneglycol
EP0307434B2 (en) 1987-03-18 1998-07-29 Scotgen Biopharmaceuticals, Inc. Altered antibodies
DE3889853D1 (de) 1987-11-05 1994-07-07 Hybritech Inc Polysaccharidmodifizierte Immunglobuline mit reduziertem immunogenem Potential oder verbesserter Pharmakokinetik.
ATE135397T1 (de) 1988-09-23 1996-03-15 Cetus Oncology Corp Zellenzuchtmedium für erhöhtes zellenwachstum, zur erhöhung der langlebigkeit und expression der produkte
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
DE3920358A1 (de) 1989-06-22 1991-01-17 Behringwerke Ag Bispezifische und oligospezifische, mono- und oligovalente antikoerperkonstrukte, ihre herstellung und verwendung
US7144731B2 (en) 1989-10-16 2006-12-05 Amgen Inc. SCF antibody compositions and methods of using the same
US5427908A (en) 1990-05-01 1995-06-27 Affymax Technologies N.V. Recombinant library screening methods
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
US5122469A (en) 1990-10-03 1992-06-16 Genentech, Inc. Method for culturing Chinese hamster ovary cells to improve production of recombinant proteins
DK0578774T3 (da) 1991-04-05 1999-04-26 Univ Washington Monoklonale antistoffer mod stamcellefaktor-receptorer
US5545533A (en) 1991-05-25 1996-08-13 Boehringer Mannheim Gmbh Monoclonal antibodies against c-kit and method of detecting a malignancy using c-kit specific antibodies
CA2122732C (en) 1991-11-25 2008-04-08 Marc D. Whitlow Multivalent antigen-binding proteins
WO1993011794A1 (en) 1991-12-13 1993-06-24 Xoma Corporation Methods and materials for preparation of modified antibody variable domains and therapeutic uses thereof
US5807549A (en) 1993-05-21 1998-09-15 Research Corporation Technologies, Inc. Lymphocyte chemoattractant factor and uses thereof
US6096871A (en) 1995-04-14 2000-08-01 Genentech, Inc. Polypeptides altered to contain an epitope from the Fc region of an IgG molecule for increased half-life
DE19600589C1 (de) 1996-01-10 1997-01-16 Univ Eberhard Karls Antikörper A3C6E2
AU728657B2 (en) 1996-03-18 2001-01-18 Board Of Regents, The University Of Texas System Immunoglobulin-like domains with increased half-lives
US6171586B1 (en) 1997-06-13 2001-01-09 Genentech, Inc. Antibody formulation
US6660843B1 (en) 1998-10-23 2003-12-09 Amgen Inc. Modified peptides as therapeutic agents
KR100483494B1 (ko) 1998-12-01 2005-04-15 프로테인 디자인 랩스 인코포레이티드 감마 인터페론에 대한 인간화 항체
JP3597140B2 (ja) 2000-05-18 2004-12-02 日本たばこ産業株式会社 副刺激伝達分子ailimに対するヒトモノクローナル抗体及びその医薬用途
JP4317010B2 (ja) 2001-07-25 2009-08-19 ピーディーエル バイオファーマ,インコーポレイティド IgG抗体の安定な凍結乾燥医薬製剤
LT1434871T (lt) 2001-09-20 2017-04-10 Immunex Corporation Heteromerinius polipeptidus ekspresuojančių ląstelių atranka
JP2007533732A (ja) 2004-04-23 2007-11-22 アブ サイエンス 線維症を処置するためのc−kit阻害剤の使用法
TWI395754B (zh) * 2006-04-24 2013-05-11 Amgen Inc 人類化之c-kit抗體

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
None *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11041022B2 (en) 2018-11-26 2021-06-22 Forty Seven, Inc. Humanized antibodies against c-Kit
US11208482B2 (en) 2018-11-26 2021-12-28 Forty Seven, Inc. Humanized antibodies against c-Kit
US12091458B2 (en) 2018-11-26 2024-09-17 Forty Seven, Inc. Humanized antibodies against c-Kit

Also Published As

Publication number Publication date
TW200808826A (en) 2008-02-16
JP5094842B2 (ja) 2012-12-12
UA95478C2 (ru) 2011-08-10
US20110223165A1 (en) 2011-09-15
JP5906120B2 (ja) 2016-04-20
JP2012126746A (ja) 2012-07-05
EA015923B1 (ru) 2011-12-30
EP3453724A1 (en) 2019-03-13
NZ572397A (en) 2011-10-28
AU2007243318A1 (en) 2007-11-08
MY153710A (en) 2015-03-13
KR20090029190A (ko) 2009-03-20
US7915391B2 (en) 2011-03-29
IL194776A0 (en) 2009-08-03
AU2007243318B2 (en) 2011-07-28
CN101472949A (zh) 2009-07-01
CA2648882A1 (en) 2007-11-08
EP2010571A2 (en) 2009-01-07
TWI395754B (zh) 2013-05-11
BRPI0712920A2 (pt) 2012-10-02
CR10444A (es) 2009-02-10
NO20084878L (no) 2008-11-19
PE20080694A1 (es) 2008-08-01
EA200802168A1 (ru) 2009-06-30
KR101110547B1 (ko) 2012-02-09
NZ594968A (en) 2013-03-28
AR060583A1 (es) 2008-06-25
IL194776A (en) 2012-10-31
US20070253951A1 (en) 2007-11-01
US20130288303A1 (en) 2013-10-31
MX2008013640A (es) 2008-11-04
WO2007127317A3 (en) 2008-02-07
WO2007127317A2 (en) 2007-11-08
ES2705480T3 (es) 2019-03-25
JP2015071649A (ja) 2015-04-16
US8436150B2 (en) 2013-05-07
JP2009534052A (ja) 2009-09-24
US8791249B2 (en) 2014-07-29
ZA200809065B (en) 2009-09-30
CA2648882C (en) 2015-10-20

Similar Documents

Publication Publication Date Title
EP2010571B1 (en) Humanized c-kit antibody
KR101125127B1 (ko) 자가면역 및 염증성 질병과 기관 이식 거부반응의 치료를위해 사용되는 길항제 항-cd40 항체의 사용 방법
JP6289375B2 (ja) 抗il−36r抗体
JP4958555B2 (ja) 安定化されたヒトIgG4抗体
KR101128777B1 (ko) 다발성 골수종을 치료하기 위한 길항제 항-cd40단클론성 항체의 용도
AU2010225951B2 (en) Pharmaceutical formulation containing improved antibody molecules
TWI431013B (zh) 有機分子
KR101352416B1 (ko) 신규의 완전한 사람형 항-vap-1 모노클로날 항체
US20130336992A1 (en) Antibodies directed to angiopoietin-1 and angiopoietin-2 and uses thereof
KR20100117108A (ko) Fc 리간드 친화성이 감소된 항-IFNAR1 항체
JP2006517191A (ja) 共刺激因子を用いた併用療法
WO2023061063A1 (zh) 靶向cd70的抗体及其制备和用途
JP2020511502A (ja) 心疾患の予防および治療方法
TWI831762B (zh) Pac1抗體及其用途

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20080925

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC MT NL PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA HR MK RS

17Q First examination report despatched

Effective date: 20100326

DAX Request for extension of the european patent (deleted)
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20180215

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAJ Information related to disapproval of communication of intention to grant by the applicant or resumption of examination proceedings by the epo deleted

Free format text: ORIGINAL CODE: EPIDOSDIGR1

GRAL Information related to payment of fee for publishing/printing deleted

Free format text: ORIGINAL CODE: EPIDOSDIGR3

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

INTC Intention to grant announced (deleted)
GRAR Information related to intention to grant a patent recorded

Free format text: ORIGINAL CODE: EPIDOSNIGR71

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE PATENT HAS BEEN GRANTED

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC MT NL PL PT RO SE SI SK TR

INTG Intention to grant announced

Effective date: 20180907

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602007056515

Country of ref document: DE

Ref country code: AT

Ref legal event code: REF

Ref document number: 1053915

Country of ref document: AT

Kind code of ref document: T

Effective date: 20181115

REG Reference to a national code

Ref country code: CH

Ref legal event code: NV

Representative=s name: PATENTANWALT MATTHIAS W. SCHAEFER, CH

REG Reference to a national code

Ref country code: NL

Ref legal event code: MP

Effective date: 20181017

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 1053915

Country of ref document: AT

Kind code of ref document: T

Effective date: 20181017

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2705480

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20190325

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: NL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181017

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181017

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190117

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181017

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181017

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181017

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181017

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190217

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190118

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190217

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181017

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602007056515

Country of ref document: DE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181017

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181017

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181017

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181017

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181017

26N No opposition filed

Effective date: 20190718

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181017

REG Reference to a national code

Ref country code: BE

Ref legal event code: MM

Effective date: 20190430

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181017

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20190424

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20190430

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181017

REG Reference to a national code

Ref country code: CH

Ref legal event code: PCAR

Free format text: NEW ADDRESS: SPLUEGENSTRASSE 10, 8002 ZUERICH (CH)

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20190424

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181017

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20181017

Ref country code: HU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT; INVALID AB INITIO

Effective date: 20070424

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20230425

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20240320

Year of fee payment: 18

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: IT

Payment date: 20240320

Year of fee payment: 18

Ref country code: FR

Payment date: 20240320

Year of fee payment: 18

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: DE

Payment date: 20240320

Year of fee payment: 18

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: CH

Payment date: 20240501

Year of fee payment: 18

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: ES

Payment date: 20240502

Year of fee payment: 18